WO2020210655A1 - Compositions virales à spécificité améliorée dans le cerveau - Google Patents

Compositions virales à spécificité améliorée dans le cerveau Download PDF

Info

Publication number
WO2020210655A1
WO2020210655A1 PCT/US2020/027708 US2020027708W WO2020210655A1 WO 2020210655 A1 WO2020210655 A1 WO 2020210655A1 US 2020027708 W US2020027708 W US 2020027708W WO 2020210655 A1 WO2020210655 A1 WO 2020210655A1
Authority
WO
WIPO (PCT)
Prior art keywords
aav capsid
aav
amino acid
cell
disease
Prior art date
Application number
PCT/US2020/027708
Other languages
English (en)
Inventor
Viviana Gradinaru
Spripriya Ravindra KUMAR
Benjamin DEVERMAN
Original Assignee
California Institute Of Technology
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by California Institute Of Technology filed Critical California Institute Of Technology
Priority to AU2020271108A priority Critical patent/AU2020271108A1/en
Priority to EP20787580.8A priority patent/EP3953371A4/fr
Priority to CN202080043549.2A priority patent/CN114127090A/zh
Priority to CA3136658A priority patent/CA3136658A1/fr
Priority to US17/602,505 priority patent/US20220220502A1/en
Priority to JP2021559829A priority patent/JP2022526018A/ja
Priority to KR1020217036564A priority patent/KR20220007056A/ko
Priority to SG11202111249YA priority patent/SG11202111249YA/en
Publication of WO2020210655A1 publication Critical patent/WO2020210655A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14143Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14151Methods of production or purification of viral material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14171Demonstrated in vivo effect

Definitions

  • rAAVs Recombinant adeno-associated viruses
  • BBB blood brain barrier
  • CNS central nervous system
  • rAAVs with engineered specificity into the capsid structure through iterative rounds of positive and negative selection, yielding variants with tropisms having an increased specificity and transduction efficiency when measured in the CNS, and in some cases, a decreased specificity and transduction efficiency in an off-target environment, like the liver.
  • the rAAVs described herein achieve widespread transduction to target environment (e.g., target cell types or tissues) in a subject upon systemic delivery (e.g., intravenous injection).
  • M-CREATE multiplexed Cre recombination-based AAV targeted evolution
  • Strategies for unbiased selection and analysis include determining variants’ enrichment score (by normalizing the target tissue library to starting virus library) and unbiased propagation between rounds of selections through a synthetic library construction (where each variant is represented equally). Also disclosed are detailed characterizations of the resultant libraries from sequencing data which provide useful insights on the selection of variants towards a target.
  • AAV capsid libraries generated using M-CREATE.
  • the first library a 7-mer peptide insertion in AAV9 ( l-mer-i library) was built for parallel in vivo selections across different brain cell types - endothelial cells, neurons, and astrocytes - and yielded a large pool of AAV9 variants with enhanced ability to target as well as cross the blood- brain barrier (BBB) and broadly transduce the central nervous system (CNS).
  • BBB blood- brain barrier
  • CNS central nervous system
  • the second library a 3-mer peptide substitution in AAV-PHP.B (3 -mer-s library), was reinvestigated by
  • a pool of AAV-PHP.B variants were discovered including a variant that transduce CNS neurons with greater specificity.
  • the rAAVs of the present disclosure can efficiently target the endothelial cells of the blood-brain barrier, variants that can broadly transduce different cell types in the central nervous system, and a variant exhibiting greater specificity towards transducing neuron.
  • AAV capsids comprising: (a) an AAV capsid protein comprising: (i) a first amino acid sequence that is at least 98% identical to amino acid 217 to amino acid 736 of SEQ ID NO: 1; and (ii) a second amino acid sequence at least 57.1% identical to an amino acid sequence provided in Tables 2-3 or FIG. 33 inserted at an amino acid position 588_589 within SEQ ID NO: 1, wherein the AAV capsid protein is characterized by at least one of an increased specificity and an increased transduction efficiency when measured in a central nervous system (CNS) in a subject when delivered to the subject systemically, relative to a native AAV capsid protein provided in SEQ ID NO: 1.
  • CNS central nervous system
  • the second amino acid sequence is at least 71.4% identical to the amino acid sequence provided in Tables 2- 3 or FIG. 33. In some embodiments, the second amino acid sequence is at least 86.7% identical to the amino acid sequence provided in Tables 2-3 or FIG. 33. In some embodiments, the second amino acid sequence is selected from the group consisting of TALKPFL, TTLKPFL, TLQIPFK, TMQKPFI, SIERPFK, RYQGDSV, and TTLKPFS.
  • the AAV capsid protein is present in VP1, VP2, and VP3 of the AAV capsid. In some embodiments, the AAV capsid is chimeric.
  • the CNS comprises a cell-type selected from the group consisting of a neuron, an oligodendrocyte, an astrocyte, and a brain vascular cell.
  • the CNS comprises a tissue that is selected from the group consisting of a brain, a thalamus, a cortex, a striatum, a ventral midbrain, and a spinal cord.
  • the AAV capsid protein further comprises an amino acid substitution A587D.
  • the AAV capsid protein further comprises an amino acid substitution Q588G.
  • the AAV capsid protein further comprises an amino acid substitution comprising A589N. In some embodiments, the AAV capsid protein further comprises an amino acid substitution comprising Q590P. In some embodiments, the second amino acid sequence at the amino acid position 588_589 within SEQ ID NO: 1 is not
  • the AAV capsid is isolated and purified.
  • the AAV capsid is formulated as a pharmaceutical formulation for intravenous administration to treat a disease or a condition of the liver, the pharmaceutical formulation further comprising a pharmaceutically acceptable carrier.
  • the pharmaceutical formulation further comprising a pharmaceutically acceptable carrier.
  • pharmaceutical formulation further comprises a therapeutic agent.
  • AAV capsids comprising an AAV capsid protein comprising a seven amino acid insertion (X1-X2-X3-X4-X5-X6-X7) between amino acid 588 and amino acid 589 in an amino acid sequence of the AAV capsid protein provided in SEQ ID NO: 1, wherein XI is an amino acid selected from the group consisting of E, D, G, R, S and T.
  • X2 is an amino acid selected from the group consisting of A, G, I, L, M, N, Q, T, and Y.
  • X3 is an amino acid selected from the group consisting of E, K, L, T, and Q.
  • X4 is an amino acid selected from the group consisting of G, I, K, L, R, T, and V.
  • X5 is an amino acid selected from the group consisting of A, D, G, P, L, Q, and V.
  • X6 is an amino acid selected from the group consisting of F, K, N, P, Q, S, and V.
  • X7 is an amino acid selected from the group consisting of I, K, L, P, and V.
  • the seven amino acid insertion is selected from the group consisting of TALKPFL, TTLKPFL, TLQIPFK, TMQKPFI, SIERPFK, RYQGDSV, and TTLKPFS.
  • the AAV capsid protein is present in VP1, VP2, and VP3 of the AAV capsid.
  • the AAV capsid is chimeric. In some embodiments, 60 copies of the AAV capsid protein are assembled into the AAV capsid.
  • the AAV capsid protein is characterized by at least one of an increased specificity and an increased transduction efficiency when measured in a central nervous system (CNS) in a subject when delivered to the subject systemically, relative to a native AAV capsid protein provided in SEQ ID NO: 1.
  • the CNS comprises a cell-type selected from the group consisting of a neuron, a glial cell, a
  • the CNS comprises a tissue that is selected from the group consisting of a brain, a thalamus, a cortex, a striatum, a ventral midbrain, and a spinal cord.
  • the AAV capsid protein further comprises an amino acid substitution comprising A587D.
  • the AAV capsid protein further comprises an amino acid substitution comprising Q588G.
  • the AAV capsid protein further comprises an amino acid substitution comprising A589N.
  • the AAV capsid protein further comprises an amino acid substitution comprising Q590P.
  • the seven amino acid insertion is not TLAVPFK, KFPVALT, SVSKPFL, FTLTTPK, MNATKNV, NGGTSSS, TRTNPEA, or YTLSQGW.
  • the AAV capsid is isolated and purified.
  • the AAV capsid is formulated as a pharmaceutical formulation for intravenous administration to treat a disease or a condition of the liver, the pharmaceutical formulation further comprising a pharmaceutically acceptable carrier.
  • the pharmaceutical formulation further comprises a therapeutic agent.
  • AAV capsids comprising: (a) an AAV capsid protein comprising: (i) a first amino acid sequence that is at least 98% identical to amino acid 217 to amino acid 736 of SEQ ID NO: 1; and (ii) a second amino acid sequence at least 57.1% identical to an amino acid sequence provided in Table 4 or FIG. 35 at an amino acid position 588_589 within SEQ ID NO: 1, wherein the AAV capsid protein is characterized by at least one of an increased specificity and an increased transduction efficiency when measured in a liver in a subject when delivered to the subject systemically, relative to a native AAV capsid protein provided in SEQ ID NO: 1.
  • the second amino acid sequence is at least 71.4% identical to the amino acid sequence provided in Table 4 or FIG. 35. In some embodiments, the second amino acid sequence is at least 86.7% identical to the amino acid sequence provided in Table 4 or FIG. 35. In some embodiments, the second amino acid sequence is selected from the group consisting of KAYSVQV, PSGSARS, and RTANALG.
  • the AAV capsid protein is present in VP1, VP2, and VP3 of the AAV capsid. In some embodiments, the AAV capsid is chimeric. In some embodiments, 60 copies of the AAV capsid protein are assembled into the AAV capsid. In some embodiments, the AAV capsid is isolated and purified.
  • the AAV capsid is formulated as a pharmaceutical formulation for intravenous administration to treat a disease or a condition of the liver, the pharmaceutical formulation further comprising a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical formulation further comprises a therapeutic agent.
  • AAV capsid proteins comprising: (i) a first amino acid sequence that is at least 98% identical to amino acid 217 to amino acid 736 of SEQ ID NO: 1; and (ii) a second amino acid sequence at least 57.1% identical to an amino acid sequence provided in Tables 2-3 or FIG. 33 inserted at an amino acid position 588_589 within SEQ ID NO: 1, wherein the AAV capsid protein is characterized by at least one of an increased specificity and an increased transduction efficiency when measured in a central nervous system (CNS) in a subject when delivered to the subject systemically, relative to a native AAV capsid protein provided in SEQ ID NO: 1.
  • CNS central nervous system
  • the second amino acid sequence is at least 71.4% identical to the amino acid sequence provided in Tables 2-3 or FIG. 33. In some embodiments, the second amino acid sequence is at least 86.7% identical to the amino acid sequence provided in Tables 2-3 or FIG. 33. In some embodiments, the second amino acid sequence is selected from the group consisting of TALKPFL, TTLKPFL, TLQIPFK, TMQKPFI, SIERPFK, RYQGDSV, and TTLKPFS.
  • the AAV capsid protein is present in VP1, VP2, and VP3 of an AAV capsid. In some embodiments, the AAV capsid is chimeric.
  • the CNS comprises a cell-type selected from the group consisting of a neuron, an oligodendrocyte, an astrocyte, and a brain vascular cell.
  • the CNS comprises a tissue that is selected from the group consisting of a brain, a thalamus, a cortex, a striatum, a ventral midbrain, and a spinal cord.
  • the AAV capsid protein further comprises an amino acid substitution A587D.
  • the AAV capsid protein further comprises an amino acid substitution Q588G.
  • the AAV capsid protein further comprises an amino acid substitution comprising A589N. In some embodiments, the AAV capsid protein further comprises an amino acid substitution comprising Q590P. In some embodiments, the second amino acid sequence at the amino acid position 588_589 within SEQ ID NO: 1 is not TLAVPFK, KFPVALT, SVSKPFL, FTLTTPK, MNATKNV, NGGTSSS, TRTNPEA, or YTLSQGW. In some embodiments, the AAV capsid protein is isolated and purified.
  • the AAV capsid protein is formulated as a pharmaceutical formulation for intravenous administration to treat a disease or a condition of the liver, the pharmaceutical formulation further comprising a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical formulation further comprises a therapeutic agent.
  • AAV capsids proteins comprising a seven amino acid insertion (X1-X2-X3-X4-X5-X6-X7) between amino acid 588 and amino acid 589 in an amino acid sequence of the AAV capsid protein provided in SEQ ID NO: 1, wherein XI is an amino acid selected from the group consisting of E, D, G, R, S and T.
  • X2 is an amino acid selected from the group consisting of A, G, I, L, M, N, Q, T, and Y.
  • X3 is an amino acid selected from the group consisting of E, K, L, T, and Q.
  • X4 is an amino acid selected from the group consisting of G, I, K, L, R, T, and V.
  • X5 is an amino acid selected from the group consisting of A, D, G, P, L, Q, and V.
  • X6 is an amino acid selected from the group consisting of F, K, N, P, Q, S, and V.
  • X7 is an amino acid selected from the group consisting of I, K, L, P, and V.
  • the seven amino acid insertion is selected from the group consisting of TALKPFL, TTLKPFL, TLQIPFK, TMQKPFI, SIERPFK, RYQGDSV, and TTLKPFS.
  • the AAV capsid protein is present in VP1, VP2, and VP3 of a AAV capsid.
  • the AAV capsid is chimeric. In some embodiments, 60 copies of the AAV capsid protein are assembled into the AAV capsid.
  • the AAV capsid protein is characterized by at least one of an increased specificity and an increased transduction efficiency when measured in a central nervous system (CNS) in a subject when delivered to the subject systemically, relative to a native AAV capsid protein provided in SEQ ID NO: 1.
  • the CNS comprises a cell-type selected from the group consisting of a neuron, a glial cell, a oligodendrocyte, an ependymal cell, an astrocyte, a Schwann cell, a satellite cell, and an enteric glial cell.
  • the CNS comprises a tissue that is selected from the group consisting of a brain, a thalamus, a cortex, a striatum, a ventral midbrain, and a spinal cord.
  • the AAV capsid protein further comprises an amino acid substitution comprising A587D.
  • the AAV capsid protein further comprises an amino acid substitution comprising Q588G.
  • the AAV capsid protein further comprises an amino acid substitution comprising A589N.
  • the AAV capsid protein further comprises an amino acid substitution comprising Q590P.
  • the seven amino acid insertion is not TLAVPFK, KFPVALT, SVSKPFL, FTLTTPK, MNATKNV, NGGTSSS, TRTNPEA, or YTLSQGW.
  • the AAV capsid protein is isolated and purified.
  • the AAV capsid protein is formulated as a pharmaceutical formulation for intravenous administration to treat a disease or a condition of the liver, the pharmaceutical formulation further comprising a pharmaceutically acceptable carrier.
  • the pharmaceutical formulation further comprises a therapeutic agent.
  • AAV capsids proteins comprising: (i) a first amino acid sequence that is at least 98% identical to amino acid 217 to amino acid 736 of SEQ ID NO: 1; and (ii) a second amino acid sequence at least 57.1% identical to an amino acid sequence provided in Table 4 or FIG. 35 at an amino acid position 588_589 within SEQ ID NO: 1, wherein the AAV capsid protein is characterized by at least one of an increased specificity and an increased transduction efficiency when measured in a liver in a subject when delivered to the subject systemically, relative to a native AAV capsid protein provided in SEQ ID NO: 1.
  • the second amino acid sequence is at least 71.4% identical to the amino acid sequence provided in Table 4 or FIG. 35. In some embodiments, the second amino acid sequence is at least 86.7% identical to the amino acid sequence provided in Table 4 or FIG. 35. In some embodiments, the second amino acid sequence is selected from the group consisting of KAYSVQV, PSGSARS, and RTANALG.
  • the AAV capsid protein is present in VP1, VP2, and VP3 of an AAV capsid. In some embodiments, the AAV capsid is chimeric. In some embodiments, 60 copies of the AAV capsid protein are assembled into the AAV capsid. In some embodiments, the AAV capsid protein is isolated and purified.
  • the AAV capsid protein is formulated as a pharmaceutical formulation for intravenous administration to treat a disease or a condition of the liver, the pharmaceutical formulation further comprising a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical formulation further comprises a therapeutic agent.
  • aspects disclosed herein comprise plasmid vectors comprising a nucleic acid sequence encoding the AAV capsids and AAV capsid proteins described herein.
  • the plasmid vector is bacterial.
  • the plasmid vector is derived from Escherichia coli.
  • the nucleic acid sequence comprises, in a 5’ to 3’ direction: (1) a 5’ inverted terminal repeat (ITR) sequence, (2) a Replication ⁇ Rep) gene, (3) a Capsid (Cap) gene, and (4) a 3’ ITR, wherein the Cap gene encodes the AAV capsid protein described herein.
  • the plasmid vector encodes a pseudotyped AAV capsid protein.
  • the Cap gene is derived from the deoxyribose nucleic acid (DNA) provided in any one of SEQ ID NOs: 6-10.
  • the nucleic acid sequence comprising the Cap gene is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to any one of the DNA sequences provided in U.S. App. Ser. No. 16/582,635, incorporated herein by reference.
  • the 5’ ITR and the 3’ ITR are derived from an AAV2 serotype.
  • the 5’ ITR and the 3’ ITR are derived from an AAV5 serotype. In some instances, the 5’ ITR and the 3’ ITR are derived from an AAV9 serotype. [0015] Aspects disclosed herein provide methods of treating a disease or condition in a subject comprising administering a therapeutically effective amount of a pharmaceutical formulation comprising the AAV capsid protein or the AAV capsid of the present disclosure.
  • the disease or the condition is a disease or a condition of a central nervous system (CNS) or a liver of the subject.
  • CNS central nervous system
  • the disease or condition of the liver is selected from the group consisting of Alagille Syndrome, Alcohol-Related Liver Disease, Alpha- 1 Antitrypsin Deficiency, Autoimmune Hepatitis, Benign Liver Tumors, Biliary Atresia, Cirrhosis, Crigler-Najjar Syndrome, Galactosemia, Gilbert Syndrome, Hemochromatosis, Hepatic Encephalopathy, Hepatitis A, Hepatitis B, Hepatitis C, Hepatorenal Syndrome,
  • the disease or condition of the CNS is selected from group consisting of Absence of the Septum Pellucidum, Acid Lipase Disease, Acid Maltase Deficiency, Acquired Epileptiform Aphasia, Acute Disseminated
  • ADHD Attention Deficit-Hyperactivity Disorder
  • Adie's Pupil Adie's Syndrome
  • Adrenoleukodystrophy Agenesis of the Corpus Callosum, Agnosia, Aicardi
  • CIDP Chronic Orthostatic Intolerance, Chronic Pain, Cockayne Syndrome Type II, Coffin Lowry Syndrome, Colpocephaly, Coma, Complex Regional Pain Syndrome, Congenital Facial Diplegia, Congenital Myasthenia, Congenital Myopathy, Congenital Vascular Cavernous Malformations, Corticobasal Degeneration, Cranial Arteritis, Craniosynostosis, Cree encephalitis, Creutzfeldt- Jakob Disease, Cumulative Trauma Disorders, Cushing's Syndrome, Cytomegalic Inclusion Body Disease, Cytomegalovirus Infection, Dancing Eyes-Dancing Feet Syndrome, Dandy-Walker Syndrome, Dawson Disease, Deafness, De Morsier's Syndrome, Dejerine-Klumpke Palsy, Dementia, Dementia -Multi -Infarct, Dementia - Semantic, Dementia - Subcortical, Dementia With Lewy
  • Encephalopathy familial infantile
  • Encephalotrigeminal Angiomatosis Epilepsy
  • Epileptic Hemiplegia Erb's Palsy, Erb-Duchenne and Dejerine-Klumpke Palsies
  • Essential Tremor Extrapontine Myelinolysis
  • Fabry Disease Fahr's Syndrome
  • Fainting Familial Dysautonomia
  • Familial Hemangioma Familial Idiopathic Basal Ganglia Calcification
  • Familial Periodic Paralyses Familial Spastic Paralysis, Farber's Disease, Febrile Seizures
  • Mucopolysaccharidoses Multi-Infarct Dementia, Multifocal Motor Neuropathy, Multiple Sclerosis, Multiple System Atrophy, Multiple System Atrophy with Orthostatic Hypotension, Muscular Dystrophy, Myasthenia -Congenital, Myasthenia Gravis, Myelinoclastic Diffuse Sclerosis, Myoclonic Encephalopathy of Infants, Myoclonus, Myopathy, Myopathy- Congenital, Myopathy -Thyrotoxic, Myotonia, Myotonia Congenita, Narcolepsy, Neuroacanthocytosis, Neurodegeneration with Brain Iron Accumulation, Neurofibromatosis, Neuroleptic Malignant Syndrome, Neurological Complications of AIDS, Neurological Complications of Lyme Disease, Neurological Consequences of Cytomegalovirus Infection, Neurological Manifestations of Pompe Disease, Neurological Sequelae Of Lupus, Neuromyelitis Optica, Neuromyotonia, Neuronal Ceroid Lipofuscinos
  • Leukoencephalopathy Progressive Sclerosing Poliodystrophy, Progressive Supranuclear Palsy, Prosopagnosia, Pseudo-Torch syndrome, Pseudotoxoplasmosis syndrome, Pseudotumor Cerebri, Psychogenic Movement, Ramsay Hunt Syndrome I, Ramsay Hunt Syndrome II, Rasmussen's Encephalitis, Reflex Sympathetic Dystrophy Syndrome, Refsum Disease, Refsum Disease - Infantile, Repetitive Motion Disorders, Repetitive Stress Injuries, Restless Legs Syndrome, Retrovirus-Associated Myelopathy, Rett Syndrome, Reye's Syndrome, Rheumatic Encephalitis, Riley-Day Syndrome, Sacral Nerve Root Cysts, Saint Vitus Dance, Salivary Gland Disease, Sandhoff Disease, Schilder's Disease, Schizencephaly, Seitelberger Disease, Seizure Disorder, Semantic Dementia, Septo-Optic Dysplasia, Se
  • SUNCT Neuralgiform
  • the pharmaceutical formulation comprises a therapeutic nucleic acid encoding a therapeutic gene expression product.
  • the therapeutic gene expression product is effective to modulate an activity or an expression of a target gene or gene expression product selected from the group consisting of Sarcoglycan Alpha (SGCA), glutamic acid decarboxylase 65 (GAD65), glutamic acid decarboxylase 67 (GAD67), CLN2, Nerve Growth Factor (NGF), Survival Of Motor Neuron 1, Telomeric (SMN1), Factor X (FIX), Retinoid Isomerohydrolase (RPE65), sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a), b-Glucocerebrosidase (GCase), Frataxin (FXN), Huntingtin (HTN), methyl-CpG binding protein 2 (MECP2), a peroxisomal biogenesis factor (PEX), progranulin (GRN), an antitubulin agent, copper-zinc superoxide dismutase (SOD1), Glucosylceramidase
  • the therapeutic gene expression product comprises gene editing components.
  • the gene editing components are selected from the group consisting of small interfering RNA (siRNA), short hairpin RNA (shRNA), a microRNA (miRNA), artificial site-specific RNA endonuclease (ASRE), zinc finger endonuclease (ZFN), CRISPR/Cas, and transcription factor like effector nuclease (TALEN).
  • aspects disclosed herein provide methods of manufacturing a recombinant AAV particle from the AAV capsid of the present disclosure, the method comprising: (a) introducing into a cell a nucleic acid comprising: (i) a first nucleic acid sequence encoding a therapeutic gene expression product; (ii) a second nucleic acid sequence encoding a recombinant viral genome comprising a capsid (Cap) gene modified to express the AAV capsid of the present disclosure; and (iii) a third nucleic acid sequence encoding an AAV helper vims genome; and (b) assembling the recombinant AAV particle, the recombinant AAV particle comprising the AAV capsid encapsidating the first nucleic acid.
  • aspects disclosed herein provide methods of manufacturing comprising: (a) introducing into a cell a nucleic acid comprising: (i) a first nucleic acid sequence encoding a therapeutic gene expression product enclosed by a 5’ and a 3’ inverted terminal repeat (ITR) sequence; (ii) a second nucleic acid sequence encoding a viral genome comprising a 5’ ITR sequence, a Replication (Rep) gene, Capsid (Cap) gene, and a 3’ ITR, wherein the Cap gene encodes the AAV capsid protein described herein; and (iii) a third nucleic acid sequence encoding a first helper vims protein selected from the group consisting of E4orf6, E2a, and VA RNA, and optionally, a second helper vims protein comprising Ela or Elb55k; (b) expressing in the cell the AAV capsid protein described herein; (c) assembling an AAV particle comprising the AAV capsi
  • the cell is mammalian. In some instances, the cell is immortalized. In some instances, the immortalized cell is an embryonic stem cell. In some instances, the embryonic stem cell is a human embryonic stem cell. In some instances, the human embryonic stem cell is a human embryonic kidney 293 (HEK-293) cell.
  • the Cap gene is derived from the deoxyribose nucleic acid (DNA) provided in any one of SEQ ID NOs: 6-10. In some instances, the nucleic acid sequence comprising the Cap gene is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to any one of the DNA sequences provided in U.S. App. Ser. No.
  • the 5’ ITR and the 3’ ITR are derived from an AAV2 serotype. In some instances, the 5’ ITR and the 3’ ITR are derived from an AAV5 serotype. In some instances, the 5’ ITR and the 3’ ITR are derived from an AAV9 serotype.
  • the first nucleic acid sequence and the second nucleic acid sequence are in trans. In some instances, the first nucleic acid sequence and the second nucleic acid sequence are in cis. In some instances, the first nucleic acid sequence, the second nucleic acid sequence and the third nucleic acid sequence, are in trans.
  • aspects disclosed herein provide methods of manufacturing a recombinant AAV particle, the method comprising: (a) providing a recombinant AAV genome comprising: (i) an AAV capsid gene, and (ii) a recognition sequence for a Cre recombinase, wherein the
  • the recognition sequence facilitates a recombinase-dependent change that is detectable, and wherein the recombinase recognition sequence comprises two Cre-recognition sites; (iii) transfecting a population of cells expressing the Cre recombinase with the recombinant AAV genome, whereby the Cre recombinase induces a recombination event to generate the recombinase- dependent change in the recombinant AAV genome, and wherein the recombinase-dependent change comprises an inversion of the sequence that is flanked by the Cre-recognition sites; (iv) detecting an increased rate of the recombinase-dependent change a target cell in the population of cells; (v) detecting a decreased rate of the recombinase-dependent change in an off-target cell in the population of cells; and (vi) identifying a recombinant AAV genome generated by the recombinase-dependent change
  • the target cell is a cell selected from the group consisting of a neuron, a glial cell, a oligodendrocyte, an ependymal cell, an astrocyte, a Schwann cell, a satellite cell, and an enteric glial cell.
  • kits comprising: (a) a first vector comprising the recombinant vector of the present disclosure; (b) a second vector encoding a helper virus protein; and (c) a third vector comprising a therapeutic nucleic acid encoding a therapeutic gene expression product.
  • kits comprising: (a) a first vector comprising a first nucleic acid sequence encoding a viral genome comprising in a 5’ to 3’ direction: (i) a 5’ inverted terminal repeat (ITR) sequence; (ii) a Replication ⁇ Rep) gene; (iii) a Capsid ⁇ Cap) gene encoding the AAV capsid proteins described herein, and (iv) a 3’ ITR; and (b) optionally, a second vector comprising a second nucleic acid sequence encoding a helper virus protein comprising at least one of E4orf6, E2a, VA RNA, Ela and Elb55k.
  • ITR inverted terminal repeat
  • a second vector comprising a second nucleic acid sequence encoding a helper virus protein comprising at least one of E4orf6, E2a, VA RNA, Ela and Elb55k.
  • the kit further comprises a cell.
  • the cell is mammalian.
  • the cell is immortalized.
  • the immortalized cell is an embryonic stem cell.
  • the embryonic stem cell is a human embryonic stem cell.
  • the human embryonic stem cell is a human embryonic kidney 293 (HEK-293) cell.
  • the kit further comprises an AAV vector comprising a heterologous nucleic acid encoding a therapeutic gene expression product.
  • the AAV vector is an episome.
  • FIG. 1 shows a multiplexed selection approach to identify capsids with specific and broad tropisms. Steps 1-6 describe the workflow in Round-1 (Rl) selection, steps 7-9 describe Round-2 (R2) selection using synthetic pool method, steps la, 2a, and 6a-b show the
  • FIG. 2 shows a structural model of the AAV9 capsid (PDB 3UX1) with the insertion site for the 7-mer-i library highlighted in red in the 60-meric (left), trimeric (middle), and monomeric (right) forms.
  • FIG. 3 shows Empirical Cumulative Distribution Frequency (ECDF) of R1 DNA and virus libraries that were recovered by deep sequencing post Gibson assembly and virus production, respectively.
  • ECDF Empirical Cumulative Distribution Frequency
  • the enrichment scores of the AAV-PHP.V2 variant are mapped as well.
  • FIG. 5 is a schematic of R2 synthetic pool (left) and PCR pool (right) library design.
  • FIG. 6 shows an overlapping bar chart showing the percentage of library overlap between the mentioned libraries and their theoretical composition.
  • FIG. 7 shows histograms of DNA and virus libraries from the two methods, where the variants in a library are binned by their read counts (in log 10 scale) and the height of the histogram is proportional to their frequency.
  • FIG. 10 shows heatmaps representing the magnitude (log2 fold change) of a given AA’s relative enrichment or depletion at each position given statistical significance is reached (boxed if P-value ⁇ 0.0001, two-sided, two-proportion z-test, p-values corrected for multiple comparisons using Bonferroni correction).
  • R2 DNA normalized to oligopool top, -9000 AA sequences
  • FIG. 12 shows clustering analysis of variants from synthetic pool brain libraries after enrichment in Tek-Cre (left), GFAP-Cre (middle), and combined SNAP-Cre and Syn-Cre (right) selections. Size of nodes represents relative enrichment in brain. Thickness of edges
  • FIG. 13 shows the 7-mer insertion peptide sequences of AAV-PHP variants between AA positions 588-589 of AAV9 capsid.
  • the images were acquired under same microscope settings as that of the sagittal sections of brain (top) with higher magnification image from cortex (bottom).
  • aGLUTl antibody staining was used in the image to show vasculature.
  • FIG. 16 shows percentage of vasculature stained with aGLUTl that overlaps with mNeongreen (XFP) expression in cortex.
  • XFP mNeongreen
  • FIG. 17 shows percentage of cells stained with each cell-type specific marker (aGLUTl, aSlOO for astrocytes, aNeuN for neurons, a01ig2 for oligodendrocyte lineage cells) that overlaps with mNeongreen (XFP) expression in cortex.
  • Kruskal-Wallis test P-value of 0.0078
  • uncorrected Dunn’s test P-value of 0.0235 for neuron vs vascular cells, and 0.0174 for neuron vs astrocyte, respectively
  • FIG 20 shows transduction by AAV-PHP.B4-B6 and Cl variants, as well as B, eB, and AAV9 controls in sagittal brain and liver sections.
  • AAV-PHP.B4-B6 and Cl variants were matched with AAV-PHP.eB.
  • the white box on the sagittal brain images marks the thalamus and not the precise region of the figures to the right.
  • Tissues are stained with cell-type specific markers: aNeuN for neurons, aSlOO for astrocytes and a01ig2 for oligodendrocyte lineage cells. Liver tissues are stained with a DNA stain, DAPI.
  • a two-way ANOVA with correction for multiple comparisons using Tukey’s test is reported with adjusted P-values (****p ⁇ 0.0001, ***P ⁇ 0.001, **P ⁇
  • the dataset comprises a mean of 2 images per region per cell-type marker per mouse).
  • FIG. 24 shows the design of the 3-mer-s PHP.B library with combinations of three AA diversification between AA 587-597 of AAV-PHP.B (or corresponding AA 587-590 of AAV9). Shared AA identity with the parent AAV-PHP.B is shown along with unique motifs for AAV-PHP.N and AAV-PHP.eB.
  • FIG. 25 shows distributions of R2 brain and liver libraries (at AA level) by enrichment score (normalized to R2 virus library, with variants sorted in decreasing order of enrichment score). The enrichment of AAV-PHP.eB and AAV-PHP.N across all libraries are mapped on the plot.
  • FIG. 26 shows heatmap represents the magnitude (log2 fold change) of a given AA’s relative enrichment or depletion at each position across the diversified region, only if statistical significance is reached on fold change (boxed if p-value ⁇ 0.0001, two-sided, two-proportion z- test, p-values corrected for multiple comparisons using Bonferroni correction).
  • FIG. 27 shows clustering analysis of enriched variants from Vgat brain library with node size representing the degree of negative enrichment in liver and the thickness of edges (connecting lines) representing degree of relatedness between nodes. Two distinct families are highlighted in yellow and their corresponding AA frequency logos are shown below (AA size represents prevalence and color encodes AA properties).
  • a two-way ANOVA with correction for multiple comparisons using Tukey’s test gave adjusted P-values reported as ****P ⁇ 0.0001, ns for P > 0.05, 95% CL
  • FIG. 30 shows clustering analysis showing the brain-enriched sequence families of all variants described herein, either identified in prior studies (PHP.B-B3, PHP.eB) or in the current study (PHP.B4-B8, PHP.V1-2, PHP.Cl-3).
  • the AA sequences inserted between 588-589 (of AAV9 capsid) for all the variants discussed are shown below.
  • FIG. 31 shows transduction of AAV9, AAV-PHP.V1 and AAV-PHP.N across three different mouse strains: C57BL/6J, BALB/cJ and FVB/NJ in sagittal brain sections (right), along with a higher magnification image of the thalamus brain region (left).
  • FIG. 32 shows transduction by AAV-PHP.B, AAV-PHP.C1-C3 in C57BL/6J and BALB/cJ mice in sagittal brain sections (right), along with a higher magnification image of the thalamus brain region (left).
  • the white box on the sagittal brain images represents the location of thalamus and not the precise area that is zoomed-in on the figure to the left.
  • the microscope settings of acquired images were matched across all sagittal sections and across all thalamus regions.
  • the insets in AAV-PHP.V1 are zoom-ins with enhanced brightness.
  • the data reported in FIGS. 31 and 32 are from one independent trial where all viruses were freshly prepared and titered in the same assay for dosage consistency.
  • FIG. 33 provides 7-mer and 11-mer variants that were positively enriched in the brain tissue in all cell lines.
  • FIG. 34 provides 7-mer and 11-mer variants enriched in one specific library, but negatively enriched in all other brain and liver libraries.
  • FIG. 35 provides 11-mer variants that were positively enriched in all ere lines in liver tissue.
  • FIG. 36 is a diagram of the genetic switch used in M-CREATE.
  • the Acceptor Vector shows the position of the forward and reverse primers between the Lox sites that are used for selective recovery of capsids from the Cre-i- cells.
  • the Rep-AAPACap vector shows a deletion of 480 bp in Cap gene in addition to the stop codons that are designed to prevent synthesis of VP1, VP2, and VP3 proteins. AAP protein translation is unaffected by these modifications.
  • FIG. 37 is a schematic of the protocol to selectively recover rAAV genomes from the target population using the Cre-Lox flipping strategy and preparation of the sample for deep sequencing.
  • FIG. 38 illustrates the library coverage for R1 DNA and virus libraries obtained from specific sequencing depths.
  • FIG. 39 shows the percentage of variant overlap within the sampled DNA and vims, or across different Cre lines within tissues, or across tissues from R1 selection.
  • FIG. 40 shows the distributions of AAV capsid read counts for libraries recovered by NGS from brain tissue across different Cre transgenic mice post R1 selection.
  • the dotted line is illustrative only and roughly separates the signal from noise (see Methods for estimation of signal v.s. noise) where signal in this context represents the input for the R2 selection.
  • FIG. 41 shows rAAV genome recovery from tissues using different treatments are shown with total rAAV genome recovery from 0.1 g of liver.
  • FIG. 42 shows the percentage of rAAV genomes recovered per ng of total extracted DNA.
  • FIG. 43 shows the CT value (cycle threshold from qPCR) of rAAV genome extracted by trizol that were treated with Smal restriction enzyme or untreated.
  • n 4 mice; 2 from GFAP-Cre line and 2 from Tek-Cre line, each data point is drawn from the mean of three technical replicates, error bar is mean+S.E.M., Mann-Whitney test, two-tailed (exact P-value of 0.0286 (*P ⁇ 0.05), in FIGS. 41, 42, and 44, and 0.1143 (n.s., P > 0.05, Cl 95%) in FIG. 43).
  • the data reported FIGS. 41, 42, and 44 are from one independent trial, and FIG. 43, from three independent trials.
  • FIG. 45 shows the vector yields obtained per 10 ng of capsid DNA library across R1 and R2 vector productions.
  • FIG. 46 shows distributions of the DNA and virus libraries produced by the synthetic pool and PCR pool methods by the standard score of NGS read counts. The variants in virus libraries are sorted by the decreasing order of standard score and their scores from respective DNA libraries are mapped onto them.
  • FIG. 48 shows distributions of capsid libraries from brain tissue of two mice used in each Cre line selection, as produced by the synthetic pool (left) and PCR pool (right) designs.
  • FIG. 51 shows the difference in enrichment score between the two codon replicates of a variant, across different brain libraries, with over 8000 variants recovered in replicates.
  • FIG. 52 shows heatmaps represent the magnitude (log2 fold change) of AA bias in “output” library 1 normalized to“input” library 2 that reach statistical significance (boxed if P- value ⁇ 0.0001, two-sided, two-proportion z-test, except in R1 DNA normalized to known NNK template where one-proportion z-test was performed, and P-values corrected for multiple comparisons using Bonferroni correction).
  • R1 DNA library normalized to NNK template top left, ⁇ 9 million sequences
  • R1 virus normalized to R1 DNA libraries bottom left, -10 million sequences
  • R2 GFAP library with enrichment score above 1.0 in brain normalized to R2 vims top right, 20 sequences,
  • R2 SNAP library with enrichment score above 1.2 normalized to R2 vims bottom right, 17 sequences
  • FIG. 53 shows clustering analysis of positively enriched variants from Tek, GFAP, and combined neuron brain libraries (SNAP and Syn) by PCR pool design, and by synthetic pool design with spike-in library are shown with size of nodes representing their relative enrichment in brain, and the thickness of edges (connecting lines) representing the extent of shared AA identity between nodes.
  • a distinct family is highlighted in yellow with the corresponding A A frequency logo below (AA size reflects prevalence and color coded based on AA properties).
  • FIG. 58 shows expression of self-complementary (sc) AAV-PHP.V1 carrying CB6 ubiquitous promoter driving EGFP (above) and CAG promoter driving EGFP (below).
  • FIGS. 56, 57, and the left column of FIG. 58 are reported from one independent trial from a fresh batch of viruses, and titered in the same assay for dosage consistency.
  • the sagittal brain images (left) were acquired using the same microscope settings to that of the sagittal brain images in FIG. 14.
  • Higher magnification images of AAV-PHP.V2 transduced brain sections stained with aGLUT or aSlOO or a01ig2 are shown.
  • One-way ANOVA, non-parametric Kruskal-Wallis test gave an approximate P-value of 0.0088).
  • FIGS. 66-67 show distributions of R1 (FIG. 66) and R2 (FIG. 67) brain libraries (at AA level, standard score (SS) of RCs sorted in decreasing order of scores).
  • the SS for AAV- PHP.N and AAV-PHP.eB across libraries are mapped on the zoomed-in view of this plot (dotted line box).
  • FIG. 68 shows a heatmap of AA distributions across the diversified region of the positively enriched variants from R2 liver library (top 100 sequences) normalized to the R2 virus (input library).
  • FIG. 69 shows clustering analysis of positively enriched variants from GFAP and Vglut2 brain libraries are shown with size of nodes representing their relative negative enrichment in liver, and the thickness of edges (connecting lines) representing their relative identity between nodes.
  • FITC-HCR Fluorescence in situ hybridization chain reaction
  • FIG. 72 shows transduction of AAV9, AAV-PHP.eB and AAV-PHP.V1 in human brain microvascular endothelial cell culture (HBMEC).
  • HBMEC human brain microvascular endothelial cell culture
  • the vectors were packaged with ssAAV:CAG-mNeongreen.
  • FIG. 73 shows the transduction of cortex brain region by AAV-PHP.B, AAV- PHP.C2 and AAV-PHP.C3 across two different mouse strains: C57BL/6J and BALB/cJ.
  • the data reported in FIGS. 72 and 73 are from one independent trial where all viruses were freshly prepared and tittered in the same assay for dosage consistency, with additional validation for AAV-PHP.C2 and AAV-PHP.C3 in an independent trial for BALB/cJ.
  • FIG. 74 provides details of a spike-in library of AAV9 and -50 additional variants (and their alternative codon duplicates), identified in prior publications (includes well characterized variants like AAV-PHP.B or AAVPHP.eB as well as many variants identified using the previous methodology but uncharacterized in vivo ) act as internal selection controls and standards for the relative performance of the new variants.
  • Deverman, B. E. et al. Cre- dependent selection yields AAV variants for widespread gene transfer to the adult brain. Nat. Biotechnol. 34, 204-209 (2016) and Chan, K. Y. et al. Engineered AAVs for efficient noninvasive gene delivery to the central and peripheral nervous systems. Nat. Neurosci. 20,
  • the spike- in library was generated as part of the synthetic pool library.
  • FIG. 75 shows enrichment scores for the spike-in library.
  • modified adeno-associated (AAV) vims capsid compositions useful for integrating a transgene into a target cell or environment (e.g ., a cell-type or tissue) in a subject when they are administered systemically (e.g., intravenous, intranasal) to the subject.
  • the modified AAV capsid proteins of the present disclosure comprise at least one insertion or substitution of an amino acid in a corresponding parental AAV capsid protein that confers a desired tropism such as an increased or decreased specificity as compared to a reference AAV capsid protein, or increased or decreased transgene transduction efficiency as compared to a reference AAV capsid protein.
  • AAV capsids engineered with desired tropisms, such as an increased specificity of viral transduction in a target in vivo environment, such as a tissue or cell.
  • the AAV capsids of the present disclosure are engineered to specifically target the central nervous system (CNS) of a subject.
  • the AAV capsids of the present disclosure are engineered to specifically target the liver of a subject.
  • the AAV capsids can encapsidate a viral vector with a heterologous nucleic acid encoding, for example, a therapeutic gene expression product.
  • heterologous nucleic acid in a target in vivo environment e.g., brain, liver
  • a subject of the AAV capsid of the present disclosure encapsidating a heterologous nucleic acid.
  • the AAV capsids disclosed herein are advantageous for many applications, such as diagnosing and/or treating monogenetic disorders of the brain (e.g., GLUT 1 -deficiency syndrome, mucoploysaccharidosis type IIIC), producing adoptive cellular therapies, and biomedical research applications.
  • the AAV capsids comprise AAV capsid proteins (e.g., VP1, VP2, and VP3), each with an insertion or a substitution of at least one amino acid at an amino acid in the 588 loop of a parental AAV capsid protein structure (AAV9 VP1 numbering).
  • AAV9 VP1 numbering The 588 loop contains the site of heparan sulfate binding of AAV2 and amenable to peptide display.
  • the only known receptors for AAV9 is N-linked terminal galactose and AAV receptor (AAVR), but many indications point toward there being others.
  • AAV9 588 loop Modifications to AAV9 588 loop are shown herein to confer an increased specificity and transgene transduction in target in vivo environments as compared to a reference AAV in rodent models.
  • the parental AAV capsid protein has an AAV serotype 9 (alias AAV9).
  • AAV-mediated transgene delivery is by direct injection to the target in vivo environment, which is disadvantageous for many reasons, including risk of injury or death, pain, and higher cost, as compared to less invasive methods.
  • intracranial injection can cause hemorrhaging of the brain.
  • Previous AAV-mediated delivery by intravenous administration avoids a need for a direct injection, but suffers from reduced specificity for the target in vivo environment (e.g., tissue or cell) resulting in off-target transduction events and necessitating a larger viral load to achieve sufficient therapeutic levels in the target in vivo environment. This is especially evident when the AAV must cross the blood brain barrier (BBB).
  • BBB blood brain barrier
  • Methods comprising systemically administering an AAV capsid of the present disclosure encapsidating a viral vector comprising a transgene (e.g ., therapeutic nucleic acid) with an increased specificity, as compared to a reference AAV capsid protein.
  • the AAV capsids of the present disclosure are capable of crossing the BBB, and transducing a transgene in a particular target cell-type (e.g., neuron, endothelial cell) in a subject.
  • the AAV capsid proteins of the present disclosure are suitable for transgene therapy to treat human disease, particularly disease that effects the target in vivo environment.
  • transgenes contained in a recombinant AAV (rAAV) vector and encapsidated by the AAV capsid proteins of the present disclosure are also provided herein.
  • the transgenes disclosed herein are delivered to a subject for a variety of purposes, such as to treat a disease or condition in the subject.
  • the transgene can be gene editing components that modulate the activity or expression of a target gene or gene expression product.
  • the transgene is a gene encoding a therapeutic gene expression product that is effective to modulate the activity or expression of itself, or another target gene or gene expression product.
  • M-CREATE multiplexed Cre recombination-based AAV targeted evolution
  • the M-CREATE method of the present disclosure supports (1) the calculation of a true enrichment score for each variant by using deep sequencing to correct for biases in viral production prior to selection, (2) reduced propagation of bias in successive rounds of selection through the creation of a post-round 1 synthetic pool library with equal variant representation, (3) the reduction of false positives by including two codon replicates of each selected variant in the pool, and (4) both positive and negative selection criteria by comparing deep sequencing of recovered capsid libraries among multiple targets (cells types or organs).
  • AAV capsids comprising the AAV capsid proteins and viral vector encoding a therapeutic nucleic acid.
  • the AAV capsid proteins are produced by introducing into a cell (e.g., immortalized stem cell) a first vector encoding the transgene (e.g ., containing the therapeutic nucleic acid), a second vector encoding the AAV genome with a AAV capsid protein, and a third vector encoding helper virus proteins, required for assembly of the AAV capsid structure and packaging of the transgene in the AAV capsid.
  • the assembled AAV capsid can be isolated and purified from the cell using suitable methods known in the art.
  • the recombinant AAV vectors comprising a nucleic acid sequence encoding the AAV capsid proteins of the present disclosure as also provided herein.
  • the viral vectors of the present disclosure comprise a nucleic acid sequence comprising the AAV viral Cap (Capsid) encoding VP1, VP2, and VP3, at least one of which is modified to produce the AAV capsid proteins of the present disclosure.
  • the recombinant AAV vector provided can be derived from an AAV serotype (e.g., AAV9).
  • Recombinant adeno-associated virus (rAAV) mediated gene delivery leverages the AAV mechanism of viral transduction for nuclear expression of an episomal heterologous nucleic acid (e.g., a transgene, therapeutic nucleic acid).
  • an episomal heterologous nucleic acid e.g., a transgene, therapeutic nucleic acid
  • a rAAV Upon delivery to a host in vivo environment, a rAAV will (1) bind or attach to cellular surface receptors on the target cell, (2) endocytose, (3) traffic to the nucleus, (4) uncoat the virus to release the encapsidated heterologous nucleic acid , (5) convert of the heterologous nucleic acid from single-stranded to double-stranded DNA as a template for transcription in the nucleus, and (6) transcribe of the episomal heterologous nucleic acid in the nucleus of the host cell (“transduction”).
  • rAAVs engineered to have an increased specificity (binding to cellular surface receptors on the target cell) and transduction efficiency (transcription of the episomal heterologous nucleic acid in the host cell) are desirable for gene therapy applications.
  • a rAAV comprises an AAV capsid that can be engineered to encapsidate a heterologous nucleic acid (e.g., therapeutic nucleic acid, gene editing machinery).
  • the AAV capsid is made up of three AAV capsid protein monomers, VP1, VP2, and VP3. Sixty copies of these three VP proteins interact in a 1:1:10 ratio to form the viral capsid (FIG. 2).
  • VP1 covers the whole of VP2 protein in addition to a -137 amino acid N-terminal region (VPlu)
  • VP2 covers the whole of VP3 in addition to -65 amino acid N-terminal region (VP1/2 common region).
  • the three capsid proteins share a conserved amino acid sequence of VP3, which in some cases is the region beginning at amino acid position 138 ( e.g ., AA139-736).
  • the AAV VP3 structure contains highly conserved regions that are common to all serotypes, a core eight- stranded b-barrel motif (bB-bI) and a small a-helix (aA).
  • the loop regions inserted between the b-strands consist of the distinctive HI loop between b-strands H and I, the DE loop between b-strands D and E, and nine variable regions (VRs), which form the top of the loops.
  • VRs such as the AA588 loop, are found on the capsid surface and can be associated with specific functional roles in the AAV life cycle including receptor binding, transduction and antigenic specificity.
  • AAV capsids comprising AAV capsid proteins with a substitution at the 588 loop that confer a desired tropism characterized by a higher specificity for transduction in specific cell-types, including for e.g., brain cell types (e.g., brain endothelial cells, neurons, astrocytes) and liver cell types.
  • the AAV capsid proteins disclosed herein enable rAAV-mediated transduction of a heterologous nucleic acid (e.g., transgene) in the brain or the liver of a subject.
  • the AAV capsids of the present disclosure, or the AAV capsid proteins may be formulated as a pharmaceutical composition.
  • the AAV capsids or the AAV capsid proteins can be isolated and purified to be used for a variety of applications.
  • recombinant AAV (rAAV) capsids comprise AAV capsid proteins that are engineered with a modified capsid protein (e.g., VP1, VP2, VP3).
  • the rAAV capsid proteins of the present disclosure are generated using the methods disclosed herein (e.g., M-CREATE).
  • the AAV capsid proteins are used in the methods of delivering a therapeutic nucleic acid (e.g., a transgene) to a subject.
  • the rAAV capsid proteins have desired AAV tropisms rendering them particularly suitable for certain therapeutic applications, e.g., the treatment of a disease or disorder in a subject such as those disclosed herein.
  • the rAAV capsid proteins are engineered for optimized entry into and through the blood brain barrier (BBB) of a subject upon systemic administration of the rAAV to the subject, such as those provided in Tables 2-3, and FIG. 33.
  • BBB blood brain barrier
  • Prior methods of AAV-mediated delivery of a therapeutic transgene to the brain required intracranial injection.
  • Intracranial injection is an invasive procedure that causes a subject discomfort, and in some cases, pain.
  • intracranial injection can cause hemorrhaging of the brain.
  • intracranial delivery has limited spread and is highly heterogeneous.
  • the rAAV capsid proteins provided in Tables 2-4, and FIG.
  • the tropisms comprise at least one of an increased specificity and efficiency (e.g ., of viral transduction) in the central nervous system (CNS) of a subject, as compared to a reference AAV.
  • the engineered AAV capsid proteins described herein have, in some cases, an insertion of an amino acid that is heterologous to the parental AAV capsid protein at the amino acid position in the 588 loop.
  • the amino acid is not endogenous to the parental AAV capsid protein at the amino acid position of the insertion.
  • the amino acid may be a naturally occurring amino acid in the same or equivalent amino acid position as the insertion of the substitution in a different AAV capsid protein.
  • amino acid insertions comprising seven amino acid polymer (7-mer) inserted at AA588_589, and may additionally include a substitution of one or two amino acids at amino acid positions flanking the 7-mer sequence (e.g., AA587-588 and/or AA589-590) to produce an eleven amino acid polymer (11-mer) at the 588 loop of a parental AAV capsid protein.
  • the 7-mers described herein were advantageously generated using polymerase chain reaction (PCR) with degenerate primers, where each of the seven amino acids is encoded by a deoxyribose nucleic acid (DNA) sequence N-N-K.“N” is any of the four DNA nucleotides and K is guanine (G) or thymine (T).
  • PCR polymerase chain reaction
  • N-N-K.“N” is any of the four DNA nucleotides and K is guanine (G) or thymine (T).
  • Recombinant AAVs were generated, each with a unique 7-mer or 11-mer at the 588 loop and each encapsidating a reporter gene that, when administered systemically in multiple transgenic animals, enabled the selective amplification and recovery of sequences that effectively transduced the reporter gene in a target in vivo environment of the transgenic animal.
  • 7-mers and 11-mers that were found to be positively enriched in the target in vivo environment (e.g, central nervous system, liver) are provided herein.“Enrichment” is the prevalence of a given 7-mer or an 11-mer in the tissue of the in vivo environment compared to its prevalence in the viral library that was administered to the transgenic animal.
  • An enrichment score above 0 indicates a positive enrichment.
  • An enrichment score below 0 indicates a negative enrichment.
  • a subset of the rAAVs with desired enrichment profiles were tested individually in vivo to determine exact systemic expression (e.g., specificity and transduction efficiency). rAAVs from this subset exhibiting a desired tropism comprising increased specificity of viral transduction, and in some cases, transduction efficiency are considered to be uniquely suited for targeted rAAV-mediated transgene delivery useful for a wide variety of purposes (e.g., therapeutic, diagnostic, scientific discovery).
  • the rAAV particles with the 7-mers or 11-mers described herein have an increased transduction efficiency in a target in vivo environment (e.g., tissue or cell type).
  • the increased transduction efficiency comprises a 1-fold, 2-fold, 3 -fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 11-fold, 12-fold, 13-fold, 14-fold, 15-fold, 16-fold, 17- fold, 18-fold, 19-fold, 20-fold, 21-fold, 22-fold, 23-fold, 24-fold, 25-fold, 26-fold, 27-fold, 28- fold, 29-fold, 30-fold, 31-fold, 32-fold, 33-fold, 34-fold, 35-fold, 36-fold, 37-fold, 38-fold, 39- fold, 40-fold, 41-fold, 42-fold, 43-fold, 44-fold, 45-fold, 46-fold, 47-fold, 48-fold, 49-fold, 50- fold, 75-fold
  • the increased transduction efficiency is at least 30-fold. In some instances, the increased transduction efficiency is at least 40-fold. In some instances, the increased transduction efficiency is at least 50-fold. In some instances, the increased transduction efficiency is at least 60-fold. In some instances, the increased transduction efficiency is at least 80-fold. In some instances, the increased transduction efficiency is at least 90-fold. In some instances, the increased transduction efficiency is at least 100-fold.
  • the rAAV particles with the 7-mers or 11-mers described herein have an increased specificity in a target in vivo environment (e.g., tissue or cell type), as compared to a reference AAV.
  • Detecting whether a rAAV possesses more or less specificity for a target in vivo environment than a reference AAV includes measuring a level of gene expression product (e.g., RNA or protein) expressed from the heterologous nucleic acid encapsidated by the rAAV in a tissue sample obtained from the target in vivo environment in a subject; and comparing the measured level to a control level (such as, for e.g., the gene expression product expressed from a heterologous nucleic acid encapsidated by a reference AAV (e.g., AAV9)).
  • a control level such as, for e.g., the gene expression product expressed from a heterologous nucleic acid encapsidated by a reference AAV (e.g
  • AAV-PHP.V2 which is shown herein to be positively enriched in the brain (enrichment score of -2.51) also exhibited an increase in reporter gene expression (e.g., measured by fluorescence reporter assay) in the brain (-60% of cortical brain vasculature cells and -60% cortical astrocytes transduced with the reporter gene) as compared to a reference AAV9 (-0%).
  • the inventors of the present disclosure would expect to see this correlation for all rAAVs disclosed herein, and further, would expect that a more significant the enrichment score (whether negative or positive) would correlate with a more significant specificity to the in vivo environment(s) as indicated by a measured level of the gene expression product in the in vivo environment(s).
  • Transduction efficiency may be measured by at least one of (1) a number of cells in a target in vivo or off-target in vivo environment expressing the heterologous nucleic acid encapsidated by the modified AAV capsid proteins disclosed herein, and (2) a quantity of expression of the heterologous nucleic acid in a single cell. Specificity for a target in vivo environment may be inferred when a presence, or an increase in a level, of rAAV-mediated transduction in a target in vivo environment is observed, as compared to a reference AAV.
  • a lack of, or reduced, specificity to an off-target in vivo environment may be inferred when an absence, or a decrease in a level, of rAAV-mediated transduction in the off-target in vivo environment is observed, as compared to a reference AAV.
  • the reference AAV may have a serotype selected from the group consisting of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11,
  • the reference AAV can have a serotype selected from the group consisting of AAV-PHP.B, AAV-PHP.eB, and AAV-PHP.S.
  • the rAAV capsid proteins of the present disclosure comprise an insertion of an amino acid in an amino acid sequence of an AAV capsid protein.
  • the AAV capsid, from which an engineered AAV capsid protein of the present disclosure is produced, is referred to as a “parental” AAV capsid.
  • the parental AAV has a serotype selected from the group consisting of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, and AAV12.
  • AAV1 is provided in GenBank Accession No. NC_002077; the complete genome of AAV-2 is provided in GenBank Accession No.
  • AAV-3 is provided in GenBank Accession No. NC_1829
  • AAV-4 is provided in GenBank Accession No. NC_001829
  • the AAV-5 genome is provided in GenBank Accession No. AF085716
  • the complete genome of AAV-6 is provided in GenBank Accession No. NC_00 1862
  • at least portions of AAV-7 and AAV-8 genomes are provided in GenBank Accession Nos. AX753246 and AX753249, respectively
  • the AAV -9 genome is provided in Gao et ah, J.
  • the parental AAV is derived from an AAV with a serotype selected from the group consisting of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, and AAV12.
  • the AAV capsid protein that is“derived” from another may be a variant AAV capsid protein.
  • a variant may include, for example, a heterologous amino acid in an amino acid sequence of the AAV capsid protein.
  • the heterologous amino acid may be non-naturally occurring in the AAV capsid protein.
  • the heterologous amino acid may be naturally occurring in a different AAV capsid protein.
  • the parental AAV capsid is described in U.S. Pat. App. Ser. Nos. 62/736,904; 16/582,635; 62/832,812; and
  • the parental AAV capsid may be modified at the 455 loop of the AAV capsid protein (e.g substitutions of 7-mer at AA452-458, AAV9 VP1 numbering).
  • the parental AAV is AAV9.
  • the amino acid sequence of the AAV9 capsid protein comprises SEQ ID NO: 1.
  • the parental AAV capsid protein sequence is 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% homologous to SEQ ID NO: 1.
  • insertions of an amino acid (or amino acid sequence) in an amino acid sequence of an AAV capsid protein at an amino acid position between amino acid 588 and amino acid 589 are insertions of an amino acid (or amino acid sequence) in an amino acid sequence of an AAV capsid protein at an amino acid position between amino acid 588 and amino acid 589.
  • “AA588_589” indicates that the insertion of the amino acid (or amino acid sequence) is immediately after an amino acid (AA) at position 588 and immediately before an AA at position 589 within an amino acid sequence of a parental AAV VP capsid protein (VP1 numbering).
  • Amino acids 587-591 include a motif comprising“AQAQA” as set forth in SEQ ID NO: 1.
  • Exemplary AAV capsid protein sequences are provided in Table 1.
  • QAVRTSL is inserted at AA588_589 in an AAV9 capsid amino acid sequence, and is provided SEQ ID NO: 3.
  • TLAVPFK is inserted at AA588_589 in an AAV9 capsid amino acid sequence, and is provided in SEQ ID NO: 2.
  • the 7- mer insertions disclosed herein may be inserted at AA588_589 in an amino acid sequence of a parental AAV9 capsid protein, a variant thereof, or equivalent amino acid position a parental AAV of a different serotype (e.g., AAV1, AAV2, AAV3, and the like).
  • the 11-mers described herein may, in some cases, comprise a 7-mer insertion at AA588_589, and substitutions of one or more amino acids at amino acid positions AA587-590. In some cases, the amino acids 587-590 are substituted with an amino acid that is not
  • AA587 is substituted with D (e.g ., A587D). In some cases, AA587 is substituted with an A ( e.g ., Q587A). In some cases, AA587 is substituted with an S (e.g., Q587S). In some cases, AA587 is substituted with a G (e.g., Q587G). In some cases, AA588 is substituted with a G (e.g., Q588G). In some cases, AA589 is substituted with an N (e.g., A589N).
  • D e.g ., A587D
  • AA587A e.g ., Q587A
  • S e.g., Q587S
  • AA587 is substituted with a G (e.g., Q587G).
  • AA588 is substituted with a G (e.g., Q588G).
  • AA589 is substituted with an
  • AA590 is substituted with a P (e.g., A590).
  • P e.g., A590.
  • SEQ ID NO: 4 comprises an insertion at AA588_AA589 of TLAVPFK, and substitutions A587D and Q588G.
  • SEQ ID NO: 5 (PHP-AAV.N) comprises an insertion at AA588_AA589 of TTLKPFS, and substitutions A587D, Q588G, A589N, and Q590P. It is envisioned that any 7-mer insertion disclosed herein in addition to a substitution with any amino acid at amino acid positions 587-590 may comprise an 11-mer.
  • the rAAV capsid proteins described herein may be isolated and purified.
  • the AAV may be isolated and purified by methods standard in the art such as by column chromatography or cesium chloride gradients. Methods for purifying AAV from helper vims are known in the art and may include methods disclosed in, for example, Clark et ah, Hum. Gene Ther., 10(6): 1031- 1039 (1999); Schenpp and Clark, Methods Mol. Med., 69: 427-443 (2002); U.S. Patent No. 6,566, 118 and WO 98/09657.
  • the rAAV capsid protein can be conjugated to a nanoparticle, a second molecule, or a viral capsid protein.
  • the nanoparticle or viral capsid protein would encapsidate the therapeutic nucleic acid described herein.
  • the second molecule is a therapeutic agent, e.g., a small molecule, antibody, antigen-binding fragment, peptide, or protein, such as those described herein.
  • the second molecule is a detectable moiety.
  • the modified AAV capsid protein conjugated to a detectable moiety may be used for in vitro , ex vivo , or in vivo biomedical research applications, the detectable moiety used to visualize the modified capsid protein.
  • the modified AAV capsid protein conjugated to a detectable moiety may also be used for diagnostic purposes.
  • AAV capsid proteins with a substitution or an insertion of at least one amino acid at an amino acid position described above in a parental AAV capsid protein that confers an increased specificity for a central nervous system (CNS) or peripheral nervous system (PNS) in a subject, even when delivered systemically.
  • CNS central nervous system
  • PNS peripheral nervous system
  • One of the many advantages of the AAV capsid proteins described herein is their ability to target the CNS and penetrate the blood brain barrier (BBB).
  • the in vivo environment can be a cell.
  • the cell can be a cell-type selected from the group consisting of a central nervous system (CNS) cell and a peripheral nervous system (PNS) cell.
  • CNS cells include a neuron and a glial cell.
  • Glial cells can be selected from the group consisting of an oligodendrocyte, an ependymal cell, and an astrocyte.
  • Non-limiting examples of a PNS cell includes a neuron or a glial cell.
  • the glial cell can be selected from the group consisting of a Schwann cell, a satellite cell, and an enteric glial cell.
  • the in vivo environment can be a tissue.
  • the tissue can be the brain, or the spinal cord.
  • the tissue can be a region of an organ, example, the cerebrum, the cerebellum, the brainstem, the cortex, the striatum, the thalamus, the lateral ventricles, the putamen, the hypothalamus, the medulla, the pons, the hippocampus, the amygdala, the motor cortex, or a combination thereof.
  • AAV capsid proteins with at least one amino acid insertion or substitution in a parental AAV capsid protein.
  • the insertion or substitution can be of at least one, two, three, four, five, six, seven, eight, nine, ten, or eleven amino acids, or more.
  • the amino acids are contiguous. In some instances, the amino acids are not contiguous.
  • the insertion is of at least one amino acid is provided in any one of the sequences provided in any one of Tables 2-3, or FIG 6. In some instances, the insertion is of at least two amino acids provided in any one of the sequences provided in any one of Tables 2-3, or FIG 6. In some instances, the insertion is of at least three amino acids provided in any one of the sequences provided in any one of Tables 2-3, or FIG 6. In some instances, the insertion is of at least four amino acids provided in any one of the sequences provided in any one of Tables 2- 3, or FIG 6. In some instances, the insertion is of at least five amino acids provided in any one of Tables 2-3, or FIG 6. In some instances, the insertion is of at least six amino acids provided in any one of Tables 2-3, or FIG 6. In some instances, the insertion is of at least seven amino acids provided in any one of Tables 2-3, or FIG 6.
  • AAV capsid proteins with an insertion of at least one amino acid XI, wherein XI is selected from the group consisting of A, E, D, G, R, S and T.
  • the insertion further comprises two amino acids, wherein X2 is selected from the group consisting of A, G, I, L, M, N, Q, R, T, and Y.
  • the insertion further comprises three amino acids, wherein X3 is selected from the group consisting of E, K, L, T, and Q.
  • the insertion further comprises at least four amino acids, wherein XI is selected from the group consisting of A, E, D, G, R, S and T, X2 is selected from the group consisting of A, G, I, L, M, N, Q, R, T, and Y, X3 is selected from the group consisting of E, K, L, T, and Q, and X4 is selected from the group consisting of G, I, K, L, M, R, T, and V. In some instances, the insertion further comprises five amino acids wherein X5 is selected from the group consisting of A, D, G, P, L, Q, and V.
  • the insertion further comprises at least six amino acids, wherein X6 is selected from the group consisting of F, K, L, N, P, Q, S, and V. In some instances, the insertion further comprises at least seven amino acids, wherein X7 is selected from the group consisting of I, K, L, P, S, and V.
  • XI, X2, X3, X4, X5, X6, and X7 are contiguous (X1-X2-X3- X4-X5-X6-X7). In some embodiments, any two of XI, X2, X3, X4, X5, X6, and X7 are contiguous. In some embodiments, any three of XI, X2, X3, X4, X5, X6, and X7 are contiguous. In some embodiments, any four of XI, X2, X3, X4, X5, X6, and X7 are contiguous. In some embodiments, any five of XI, X2, X3, X4, X5, X6, and X7 are contiguous. In some embodiments, any five of XI, X2, X3, X4, X5, X6, and X7 are contiguous. In some embodiments, any five of XI, X2, X3, X4, X5,
  • any six of XI, X2, X3, X4, X5, X6, and X7 are contiguous.
  • any seven of XI, X2, X3, X4, X5, X6, and X7 are contiguous. In some embodiments, XI, X2, X3, X4, X5, X6, and X7 are not contiguous. In some embodiments, the insertion is not TLAVPFK.
  • a 7-mer (X1-X2- X3-X4-X5-X6-X7) may in some cases advantageously have a T in position XI, an F in position X2, a P in positive X5, an F in position X6, and a K or F in position X7.
  • the 7-mer comprises T-F-X3-X4-P-F-K, wherein X3 and X4 are any amino acid.
  • the 7-mer comprises T-F-X3-X4-P-F-F, wherein X3 and X4 are any amino acid.
  • X3 is not an A. In some cases, X3 is A, S, Q, or E or F. In some cases, X4 is not a V. In some cases, X4 is R, K, V, or a Q.
  • the 7-mer may be Xl-F-A-V-P-F-K, wherein XI is any amino acid other than T, S, or N; X1-X2-A-V-P-F-K, wherein XI is any amino acid other than T, S, or N, and X2 is any amino acid other than F or V; or X1-X2-X3-V-P-F-K, wherein XI is any amino acid other than T, S, or N, X2 is any amino acid other than F or V, and X3 is any amino acid other than A, S, Q, P, or T; or X1-X2-X3-X4-P-F-K, wherein XI is any amino acid other than T,
  • X2 is any amino acid other than F or V
  • X3 is any amino acid other than A, S, Q, P, or
  • T, and X4 is any amino acid other than V, T, Q, N, F, or M.
  • the 7-mer is T-F- A-X4-P-F-K, wherein X is any amino acid other than V.
  • the 7-mer is T-L-A- X4-P-F-K, wherein X is any amino acid other than, T, Q, N, L, or M.
  • the 7-mer (X1-X2-X3-X4-X5-X6-X7) comprises TALKPFL. In some instances, the 7-mer comprises TTLKPFL. In some instances, the 7-mer comprises TLQIPFK. In some instances, the 7-mer comprises TMQKPFI. In some instances, the 7-mer comprises SIERPFK. In some instances, the 7-mer comprises RYQGDSV.
  • the AAV capsid protein comprises an insertion of at least or about three, four, five, six, or seven amino acids of an amino acid sequence T-X2-L-K-P-F-L at an amino acid position 588_589 in a parental AAV9 capsid protein (SEQ ID NO: 1), wherein X2 is A or T.
  • SEQ ID NO: 1 a parental AAV9 capsid protein
  • the AAV capsid protein has an increased specificity for viral transduction in brain vascular cells (GLUT1+), as compared to a reference AAV (e.g ., AAV9).
  • the AAV capsid protein has increased specificity for viral tranduction in astrocytes, as compared to a reference AAV (e.g., AAV9).
  • the AAV capsid protein comprises an insertion of at least or about three, four, five, six, or seven amino acids of an amino acid sequence T-X2-Q-X4-P-F-X7 at an amino acid position 588_589 in a parental AAV9 capsid protein (SEQ ID NO: 1), wherein X2 is L or M; X4 is I, K, or L; X7 is K or I.
  • the AAV capsid protein has increased specificity for viral transduction in neurons and astrocytes, as compared to a reference AAV (e.g., AAV9).
  • the amino acid sequence is TLQIPFK.
  • the amino acid sequence is TMQKPFI.
  • the amino acid sequence is TLQLPFK.
  • the AAV capsid protein comprises an insertion of at least or about three, four, five, six, or seven amino acids of an amino acid sequence S-I-E-R-P-F-K at an amino acid position 588_589 in a parental AAV9 capsid protein (SEQ ID NO: 1).
  • the AAV capsid protein has increased specificity for viral transduction in neurons and astrocytes, as compared to a reference AAV (e.g., AAV9).
  • the AAV capsid protein comprises an insertion of at least or about three, four, five, six, or seven amino acids of an amino acid sequence R-Y-Q-G-D-S-V at an amino acid position 588_589 in a parental AAV9 capsid protein (SEQ ID NO: 1).
  • the AAV capsid protein has increased specificity for viral transduction in astrocytes, as compared to a reference AAV (e.g., AAV9).
  • TABLE 2 List of 7-mer targeting peptides that can target the CNS with greater efficiency and specificity
  • AAV capsid proteins with a substitution or an insertion of at least one amino acid at an amino acid position described above in a parental AAV capsid protein that confers an increased specificity for the liver.
  • the insertion comprises at least one amino acid provided in any one of the sequences provided in Table 4 and/or FIG. 35. In some instances, the insertion comprises at least two amino acids provided in any one of the sequences provided in Table 4 and/or FIG. 35. In some instances, the insertion comprises at least three amino acids provided in any one of the sequences provided in Table 4 and/or FIG.
  • the insertion comprises at least four amino acids provided in any one of the sequences provided in Table 4 and/or FIG. 35. In some instances, the insertion comprises at least five amino acids provided in Table 4 and/or FIG. 35. In some instances, the insertion comprises at least six amino acids provided in Table 4 and/or FIG. 35. In some instances, the insertion comprises at least seven amino acids provided in Table 4 and/or FIG. 35. In some instances, the amino acids are contiguous. In some instances, the amino acids are not contiguous. In some instances, the insertion is at an amino acid position 588_589 in a parental AAV capsid protein. In some instances, the parental capsid protein is AAV9 capsid protein, provided in SEQ ID NO: 1.
  • the AAV capsids and AAV capsid proteins disclosed herein are isolated. In some instances, the AAV capsids and AAV capsid proteins disclosed herein are isolated and purified. In addition, the AAV capsids and AAV capsid proteins disclosed herein, either isolated and purified, or not, may be formulated into a pharmaceutical formulation, which in some cases, further comprises a pharmaceutically acceptable carrier.
  • the therapeutic nucleic acids useful for the treatment or prevention of a disease or condition, or symptom of the disease or condition, disclosed herein.
  • the therapeutic nucleic acids encode a therapeutic gene expression product.
  • gene expression products include proteins, polypeptides, peptides, enzymes, antibodies, antigen binding fragments, nucleic acid (RNA, DNA, antisense oligonucleotide, siRNA, and the like), and gene editing components, for use in the treatment, prophylaxis, and/or amelioration of the disease or disorder, or symptoms of the disease or disorder.
  • the therapeutic nucleic acids are placed in an organism, cell, tissue or organ of a subject by way of a rAAV, such as those disclosed herein.
  • rAAVs each comprising a viral vector (e.g., a single stranded DNA molecule (ssDNA)).
  • the viral vector comprises two inverted terminal repeat (ITR) sequences that are about 145 bases each, flanking a transgene.
  • ITR inverted terminal repeat
  • the transgene comprises a therapeutic nucleic acid, and in some cases, a promoter in cis with the therapeutic nucleic acid in an open reading frame (ORF).
  • the promoter is capable of initiating transcription of therapeutic nucleic acid in the nucleus of the target cell.
  • the ITR sequences can be from any AAV serotype. Non-limiting examples of AAV serotypes include AVI, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, and AAV12. In some cases, an ITR is from AAV2. In some cases, an ITR is from AAV9.
  • transgenes that can comprise any number of nucleotides.
  • a transgene can comprise less than about 100 nucleotides.
  • a transgene can comprise at least about 100 nucleotides.
  • a transgene can comprise at least about 200 nucleotides.
  • a transgene can comprise at least about 300 nucleotides.
  • a transgene can comprise at least about 400 nucleotides.
  • a transgene can comprise at least about 500 nucleotides.
  • a transgene can comprise at least about 1000 nucleotides.
  • a transgene can comprise at least about 5000 nucleotides. In some cases, a transgene can comprise at least about 10,000 nucleotides. In some cases, a transgene can comprise at least about 20,000 nucleotides. In some cases, a transgene can comprise at least about 30,000 nucleotides. In some cases, a transgene can comprise at least about 40,000 nucleotides. In some cases, a transgene can comprise at least about 50,000 nucleotides. In some cases, a transgene can comprise between about 500 and about 5000 nucleotides. In some cases, a transgene can comprise between about 5000 and about 10,000 nucleotides. In any of the cases disclosed herein, the transgene can comprise DNA, RNA, or a hybrid of DNA and RNA. In some cases, the transgene can be single stranded. In some cases, the transgene can be double stranded.
  • transgenes useful for modulating the expression or activity of a target gene or gene expression product thereof are encapsidated by an rAAV capsid protein of an rAAV particle described herein.
  • the rAAV particle is delivered to a subject to treat a disease or condition disclosed herein in the subject.
  • the delivery is systemic (e.g ., intravenous, intranasal).
  • transgenes disclosed herein are useful for expressing an endogenous gene at a level similar to that of a healthy or normal individual. This is particularly useful in the treatment of a disease or condition related to the underexpression, or lack of expression, of a gene expression product.
  • the transgenes disclosed herein are useful for overexpressing an endogenous gene, such that an expression level of the endogenous gene is above the expression level of a healthy or normal individual.
  • transgenes can be used to express exogenous genes (e.g., active agent such as an antibody, peptide, nucleic acid, or gene editing components).
  • the therapeutic gene expression product is capable of altering, enhancing, increasing, or inducing the activity of one or more endogenous biological processes in the cell.
  • the transgenes disclosed herein are useful for reducing expressing an endogenous gene, example, a dominant negative gene.
  • the therapeutic gene expression product is capable of altering, inhibiting, reducing, preventing, eliminating, or impairing the activity of one or more endogenous biological processes in the cell.
  • the increase of gene expression refers to an increase by at least about 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95% and 100%.
  • the protein product of the targeted gene may be increased by at least about 20%, 30%, 40%,
  • the decrease of gene expression refers to an increase by at least about 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95% and 100%.
  • the protein product of the targeted gene may be decreased by at least about 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95% and 100%.
  • endogenous sequences endogenous or part of a transgene
  • the endogenous sequences can be full-length sequences (wild-type or mutant) or partial sequences.
  • the endogenous sequences can be functional. Non-limiting examples of the function of these full length or partial sequences include increasing the serum half-life of the polypeptide expressed by a transgene (e.g., therapeutic gene) and/or acting as a carrier.
  • a transgene can be inserted into an endogenous gene such that all, some or none of the endogenous gene is expressed.
  • a transgene as described herein can be inserted into an endogenous locus such that some (N-terminal and/or C-terminal to a transgene) or none of the endogenous sequences are expressed, for example as a fusion with a transgene.
  • a transgene e.g ., with or without additional coding sequences of the endogenous gene
  • FXN Frataxin
  • a transgene can be inserted into any gene, e.g., the genes as described herein.
  • the therapeutic gene expression product is a therapeutic protein or a peptide (e.g., antibody, antigen -binding fragment, peptide, or protein).
  • the protein encoded by the therapeutic nucleic acid is between 50-5000 amino acids in length. In some embodiments the protein encoded is between 50-2000 amino acids in length. In some embodiments the protein encoded is between 50-1000 amino acids in length. In some embodiments the protein encoded is between 50-1500 amino acids in length. In some embodiments the protein encoded is between 50-800 amino acids in length. In some embodiments the protein encoded is between 50-600 amino acids in length.
  • the protein encoded is between 50-400 amino acids in length. In some embodiments the protein encoded is between 50-200 amino acids in length. In some embodiments the protein encoded is between 50-100 amino acids in length. In some embodiments the peptide encoded is between 4-50 amino acids in length. In some embodiments, the protein encoded is a tetrapeptide, a pentapeptide, a hexapeptide, a heptapeptide, an octapeptide, a nonapeptide, or a decapeptide. In some embodiments, the protein encoded comprises a peptide of 2-30 amino acids, such as for example 5-30, 10-30, 2-25, 5-25, 10-25, or 10-20 amino acids.
  • the protein encoded comprises a peptide of at least 11, 12, 13, 14, 15, 17, 20, 25 or 30 amino acids, or a peptide that is no longer than 50 amino acids, e.g. no longer than 35, 30, 25, 20, 17, 15, 14, 13, 12, 11 or 10 amino acids.
  • Non-limiting examples of therapeutic protein or peptides include an adrenergic agonist, an anti-apoptosis factor, an apoptosis inhibitor, a cytokine receptor, a cytokine, a cytotoxin, an erythropoietic agent, a glutamic acid decarboxylase, a glycoprotein, a growth factor, a growth factor receptor, a hormone, a hormone receptor, an interferon, an interleukin, an interleukin receptor, a kinase, a kinase inhibitor, a nerve growth factor, a netrin, a neuroactive peptide, a neuroactive peptide receptor, a neurogenic factor, a neurogenic factor receptor, a neuropilin, a neurotrophic factor, a neurotrophin, a neurotrophin receptor, an N-methyl-D- aspartate antagonist, a plexin, a protease, a protease inhibitor, a
  • the therapeutic protein or peptide is selected from the group consisting of brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), macrophage colony- stimulating factor (CSF), epidermal growth factor (EGF), fibroblast growth factor (FGF), gonadotropin, interferon-gamma (IFN), insulin-like growth factor 1 (IFG-1), nerve growth factor (NGF), platelet-derived growth factor (PDGF), pigment epithelium-derived factor (PEDF), transforming growth factor (TGF), transforming growth factor-beta (TGF-B), tumor necrosis factor (TNF), vascular endothelial growth factor (VEGF), prolactin, somatotropin, X-linked inhibitor of apoptosis protein 1 (XIAP1), interleukin 1 (IL-1), IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-10, viral IL
  • a therapeutic gene expression product can comprise gene editing components.
  • gene editing components include those required for CRISPR/Cas, artificial site-specific RNA endonuclease (ASRE), zinc finger endonuclease (ZFN), and transcription factor like effector nuclease (TALEN).
  • ASRE artificial site-specific RNA endonuclease
  • ZFN zinc finger endonuclease
  • TALEN transcription factor like effector nuclease
  • a subject having Huntington's disease is identified. The subject is then systemically administered a first amount of a rAAV encapsidating a viral vector encoding ZFN engineered to represses the transcription of the Huntingtin (HTT) gene.
  • the route of administration is intravenous.
  • the rAAV will include a modified AAV capsid protein that includes an amino acid sequence provided in any one of Tables 2-3, or FIG. 33, so as to allow proper targeting of the ZFN to the nervous system, while retargeting off-target organs, such as the liver. If needed, the subject is
  • a subject with cystic fibrosis is identified.
  • the subject is then systemically administered a first amount of a rAAV encapsidating a viral vector encoding ZFN engineered to represses the transcription of the cystic fibrosis transmembrane conductance regulator (CFTR) gene.
  • the route of administration is intranasal (e.g ., intranasal spray).
  • the rAAV will include a modified AAV capsid protein that includes an amino acid sequence provided in FIG. 33 or Tables 2-3, so as to allow proper targeting of the ZFN to the lung. If needed, the subject is administered a second or third dose of the rAAV, until a therapeutically effective amount of the ZFN is expressed in the subject’s lung.
  • a therapeutic nucleic acid can comprise a non-protein coding gene e.g., sequences encoding antisense RNAs, RNAi, shRNAs and micro RNAs (miRNAs), miRNA sponges or decoys, recombinase delivery for conditional gene deletion, conditional (recombinase- dependent) expression, includes those required for the gene editing components described herein.
  • the non-protein coding gene may also encode a tRNA, rRNA, tmRNA, piRNA, double stranded RNA, snRNA, snoRNA, and/or long non-coding RNA (IncRNA).
  • the non-protein coding gene can modulate the expression or the activity of a target gene or gene expression product.
  • the RNAs described herein may be used to inhibit gene expression in a target cell, for example, a cell in the central nervous system (CNS) or peripheral organ (e.g., lung).
  • inhibition of gene expression refers to an inhibition by at least about 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95% and 100%.
  • the protein product of the targeted gene may be inhibited by at least about 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95% and 100%.
  • the gene can be either a wild type gene or a gene with at least one mutation.
  • the targeted protein may be either a wild type protein or a protein with at least one mutation.
  • a therapeutic nucleic acid can modulate the expression or activity of a gene or gene expression product expressed from the gene that is implicated in disease or disorder of the brain.
  • the therapeutic nucleic acid in some cases is a gene or a modified version of the gene described herein.
  • the therapeutic nucleic acid comprises an effector gene expression product such as a gene editing component specific to target a gene therein.
  • Non limited examples of genes include Sarcoglycan Alpha (SGCA), glutamic acid decarboxylase 65 (GAD65), glutamic acid decarboxylase 67 (GAD67), CLN2 gene, Nerve Growth Factor (NGF), glial cell derived neurotrophic factor(GDNF), Neurturin, Survival Of Motor Neuron 1,
  • the peroxisomal biogenesis factor is selected from the group consisting of PEX1, PEX2, PEX3, PEX4, PEX5, PEX6, PEX7, PEX10, RECI Ib, PEX 12, PEX13, PEX14, PEX 16, PEX 19, and PEX26.
  • the gene or gene expression product is inhibited. In some instances, the gene or gene expression product is enhanced.
  • a therapeutic nucleic acid modulates expression or activity of a gene or gene expression product expressed from the gene that is implicated in disease or disorder of a particular organ (e.g., lung, heart, liver, muscle, eye).
  • a gene or gene expression product expressed from the gene that is implicated in disease or disorder of a particular organ (e.g., lung, heart, liver, muscle, eye).
  • Non-limited examples of genes include Cystic Fibrosis Transmembrane Conductance Regulator (CFTR), Factor X (FIX), RPE65, Retinoid Isomerohydrolase (RPE65), Sarcoglycan Alpha (SGCA), and sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a).
  • the therapeutic gene expression product is of human, murine, avian, porcine, bovine, ovine, feline, canine, equine, epine, caprine, lupine or primate origin. In some instances, the gene or gene expression product is inhibited. In some instances, the gene or gene expression product is enhanced.
  • AAV vectors comprising genetic information.
  • AAV vectors described herein are useful for the assembly of a rAAV and viral packaging of a heterologous nucleic acid.
  • an AAV vector may encode a transgene comprising the heterologous nucleic acid.
  • the AAV vector is from an AAV serotypes selected from the group consisting of AVI, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV11, and AAV12.
  • the AAV vector is selected from a modified AAV serotype selected from the group consisting of AAV-PHP.B, AAV-PHP.eB, and AAV-PHP.S.
  • An AAV vector can comprise a transgene, which in some cases encodes a heterologous gene expression product (e.g., therapeutic gene expression product, recombinant capsid protein, and the like).
  • the transgene is in cis with two inverted terminal repeats (ITRs) flanking the transgene.
  • the transgene may comprise a therapeutic nucleic acid encoding a therapeutic gene expression product. Due to the limited packaging capacity of the rAAV
  • transgene may be split between two AAV vectors, the first with 3’ splice donor and the second with a 5’ splice acceptor.
  • concatemers form, which are spliced together to express a full-length transgene.
  • a transgene is generally inserted so that its expression is driven by the endogenous promoter at the integration site, namely the promoter that drives expression of the endogenous gene into which a transgene is inserted.
  • a transgene comprises a promoter and/or enhancer, for example a constitutive promoter or an inducible or tissue/cell specific promoter.
  • the promoter may be CMV promoter, a CMV-P-Actin- intron-P-Globin hybrid promoter (CAG), CBA promoter, FRDA or FXN promoter, UBC promoter, GUSB promoter, NSE promoter, Synapsin promoter, MeCP2 promoter, GFAP promoter, HI promoter, U6 promoter, NFL promoter, NFH promoter, SCN8A promoter, or PGK promoter.
  • CAG CMV-P-Actin- intron-P-Globin hybrid promoter
  • promoters can be tissue- specific expression elements include, but are not limited to, human elongation factor la-subunit (EFla), immediate-early cytomegalovirus (CMV), chicken b-actin (CBA) and its derivative CAG, the b glucuronidase (GUSB), and ubiquitin C (UBC).
  • EFla human elongation factor la-subunit
  • CMV immediate-early cytomegalovirus
  • CBA chicken b-actin
  • GUSB b glucuronidase
  • UPC ubiquitin C
  • the transgene may include a tissue-specific expression elements for neurons such as, but not limited to, neuron-specific enolase (NSE), platelet-derived growth factor (PDGF), platelet-derived growth factor B-chain (PDGF-b), the synapsin (Syn), the methyl-CpG binding protein 2 (MeCP2), Ca2+/calmodulin-dependent protein kinase II
  • NSE neuron- specific enolase
  • PDGF platelet-derived growth factor
  • PDGF-b platelet-derived growth factor B-chain
  • Syn synapsin
  • the transgene may comprise a tissue-specific expression element for astrocytes such as, but not limited to, the glial fibrillary acidic protein (GFAP) and EAAT2 promoters.
  • the transgene may comprise tissue-specific expression elements for oligodendrocytes such as, but not limited to, the myelin basic protein (MBP) promoter.
  • the promoter is less than 1 kb.
  • the promoter may have a length of 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 610, 620, 630, 640, 650, 660, 670, 680, 690, 700, 710, 720, 730, 740, 750, 760, 770, 780, 790, 800 or more than 800.
  • the promoter may have a length between 200- 300, 200-400, 200-500, 200-600, 200-700, 200-800, 300-400, 300-500, 300-600, 300-700, 300- 800, 400-500, 400-600, 400-700, 400-800, 500-600, 500-700, 500-800, 600-700, 600-800 or 700-800.
  • the promoter may provide expression of the therapeutic gene expression product for a period of time in targeted tissues such as, but not limited to, the central nervous system and peripheral organs ( e.g ., lung).
  • Expression of the therapeutic gene expression product may be for a period of 1 hour, 2, hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, 18 hours, 19 hours, 20 hours, 21 hours, 22 hours, 23 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 1 week, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 2 weeks, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 3 weeks, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, 28 days, 29 days, 30 days, 31 days, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 13 months, 14 months, 15 months, 16 months, 17 months, 18 months, 19 months, 20 months, 21 months, 22 months, 23 months, 2 years, 3 years, 4 years,
  • Expression of the payload may be for 1-5 hours, 1-12 hours, 1-2 days, 1-5 days, 1-2 weeks, 1-3 weeks, 1-4 weeks, 1-2 months, 1-4 months, 1-6 months, 2-6 months, 3-6 months, 3-9 months, 4-8 months, 6-12 months, 1-2 years, 1-5 years, 2-5 years, 3-6 years, 3-8 years, 4-8 years or 5-10 years or 10-15 years, or 15-20 years, or 20-25 years, or 25-30 years, or 30-35 years, or 35-40 years, or 40-45 years, or 45-50 years, or 50-55 years, or 55-60 years, or 60- 65 years.
  • An AAV vector can comprise a genome of a helper virus.
  • Helper virus proteins are required for the assembly of a recombinant AAV (rAAV), and packaging of a transgene containing a heterologous nucleic acid into the rAAV.
  • the helper vims genes are adenovirus genes E4, E2a and VA, that when expressed in the cell, assist with AAV replication.
  • an AAV vector comprises E2.
  • an AAV vector comprises E4.
  • an AAV vector comprises VA.
  • the AAV vector comprises one of helper virus proteins, or any combination.
  • An AAV vector can comprise a viral genome comprising a nucleic acid encoding the recombinant AAV (rAAV) capsid protein described herein.
  • the viral genome can comprise a Replication (Rep) gene encoding a Rep protein, and Capsid (Cap) gene encoding an AAP protein in the first open reading frame (ORF1) or a Cap protein in the second open reading frame (ORF2).
  • the Rep protein is selected from the group consisting of Rep78, Rep68, Rep52, and Rep40.
  • the Cap gene is modified encoding a modified AAV capsid protein described herein.
  • a wild-type Cap gene encodes three proteins, VP1, VP2, and VP3. In some cases, VP1 is modified.
  • VP2 is modified.
  • VP3 is modified.
  • all three VP1-VP3 are modified.
  • the AAV vector can comprise nucleic acids encoding wild-type Rep78, Rep68, Rep52, Rep40 and AAP proteins.
  • AAV vectors comprising any one of SEQ ID NOS: 10-434, 860- 863, 868-949, 1068-5661, 14841-14880, and 14961-15053 which are the DNA sequences encoding modified portions of AAV capsid proteins of the present disclosure.
  • the AAV vector comprises a nucleic acid sequences provided in any one of SEQ ID NOS: 10- 434 and 868-949, encoding 7-mer modified AAV capsid protein portions.
  • the AAV vector of the present disclosure can comprise the VP1 Cap gene comprises any one of SEQ ID NOS: 6-9 provided in Table 5.
  • An AAV vector can comprise 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% of any one of SEQ ID NOS: 6-9.
  • the AAV9 VP1 gene provided in SEQ ID NO: 6, which is provided in Table 5, is modified to include any one of SEQ ID NOS: 10-434, 860-863, 868-949, 1068-5661, 14841-14880, and 14961-15053.
  • the AAV-PHP.eB VP1 (SEQ ID NO: 9), which is also provided in Table 5 is modified to include any one of SEQ ID NOS: 10- 434, 860-863, 868-949, 1068-5661, 14841-14880, and 14961-15053.
  • the AAV vector described herein may be used to produce a variant AAV capsid by the methods described herein.
  • FIG. 1 provides a workflow using M-CREATE.
  • M-CREATE uses an rAAV capsid genome (rAAV- Cap-in-cis-lox or rAAV-ACap-in-cis-lox2 as described in Example 2 below) that couples a full- length AAV Cap gene, controlled by regulatory elements from the AAV Rep gene, with a Cre- invertible switch.
  • the rAAV-ACap-in-cis-lox2 backbone has a bi-directional polyA flanked by two Lox sites (lox71 and lox66).
  • the ACap backbone is nonfunctional as it is missing a portion of the capsid gene.
  • Cre-Lox recombination facilitates inversion of the polyA in addition to flipping the Lox sites to Lox72 and LoxP.
  • the randomized 7-mer and 1 l-mers disclosed herein are generated using PCR, which are then inserted into the rAAV-ACap-in-cis-lox to generate vims libraries with randomized insertions or substitutions in the capsid protein sequence (e.g., at the AA588_589). (FIG. 1).
  • the vims libraries are injected into the blood stream (e.g., intravenously) of transgenic animals expressing Cre recombinase. Tissues obtained from the transgenic animals following injection in in vivo environment (e.g., brain/liver).
  • the inverted reporter expression cassette is detected using selective amplification expression of the reporter gene expression product.
  • the rAAVs in the tissues are isolated and the viral genome around the insertion site is sequenced and aligned with an AAV9 template DNA fragment.
  • the 7-mers and 1 l-mers that were recovered were enriched in the target in vivo environment, and were cloned into another rAAV-ACap-in-cis-lox2 backbone, and another round of in vivo selection is performed.
  • the 7-mers and 1 l-mers enriched in the target in vivo environment, and negatively enriched in an off-target in vivo environment are sequenced using suitable methods, such as next generation sequencing.
  • FIGS. 33-35 provide DNA sequences identified using the methods provided herein.
  • Methods comprise providing a rAAV genome comprising an AAV capsid gene, and a recognition sequence for a Cre recombinase.
  • the rAAV genome has two recognition sequences for Cre recombinase that flank a reporter expression cassette.
  • the recognition sequences for Cre recombinase e.g ., LoxP
  • Methods comprise transfecting a population of cells expressing the Cre recombinase with the rAAV genome.
  • the Cre recombinase induces an inversion (e.g.,“flip” of the reporter gene into a genome of the transgenic animal).
  • the rate of inversion e.g., a level of expression of the reporter gene in a target cell
  • the level of expression of the reporter gene is compared to a reference value, which in some cases is the rate of inversion by a reference AAV (e.g., AAV9).
  • AAV e.g., AAV9
  • a decrease in a rate of inversion is detected in an off-target cell, as compared to the rate of inversion of a reference AAV.
  • the rAAV genome recovered using the methods described herein encodes for an AAV capsid particle (e.g., capsid protein, capsid) with an increased specificity for the target cell, and a decreased specificity for the off-target cell.
  • AAV capsid particle e.g., capsid protein, capsid
  • rAAV rAAV
  • all elements that are required for AAV production in target cell are transiently transfected into the target cell using suitable methods known in the art.
  • the rAAV of the present disclosure can be product by co-transfecting three plasmid vectors, a first vector with ITR-containing plasmid carrying the transgene (e.g., therapeutic nucleic acid), a second vector that carries the AAV Rep and Cap genes; and (3), a third vector that provides the helper genes isolated from adenovirus.
  • the methods described herein generate high-titer AAV vectors that are free of adenovirus.
  • the Cap gene disclosed here comprises any one of SEQ ID NOS: 10-434, 860-863, 868-949, 1068-5661, 14841-14880, and 14961-15053 which are DNA sequences encoding the modified AAV capsid protein portions of the present disclosure.
  • rAAVs of the present disclosure are generated using the methods described in Challis, R. C. et al. Systemic AAV vectors for widespread and targeted gene delivery in rodents. Nat. Protoc. 14, 379 (2019), which is hereby incorporated by reference in its entirety. Briefly, triple transfection of HEK293T cells (ATCC) using polyethylenimine (PEI) is performed, viruses are collected after 120 hours from both cell lysates and media, and purified over iodixanol.
  • ATCC HEK293T cells
  • PEI polyethylenimine
  • a nucleic acid comprising: (1) a first nucleic acid sequence encoding a therapeutic gene expression product; (2) a second nucleic acid sequence encoding viral genome components comprising: (i) a Replication ⁇ Rep) gene encoding a Rep protein; and (ii) a modified capsid (Cap) gene encoding a modified AAV capsid protein described herein, and (3) a third nucleic acid sequence encoding a genome of an AAV helper virus; and (b) assembling a recombinant AAV (rAAV) capsid encapsidating the first nucleic acid, the rAAV capsid comprising a tropism with an increased specificity for a target in vivo environment in a subject and a decreased specificity for an off-target in vivo environment, relative to a tropism of a corresponding parental AAV capsid protein.
  • rAAV recombinant AAV
  • the methods further comprise packing the first nucleic acid sequence encoding the therapeutic gene expression product such that it becomes encapsidated by the modified AAV capsid protein.
  • the rAAV particles are isolated, concentrated, and purified using suitable viral purification methods, such as those described herein.
  • the rAAVs are generated by triple transfection of precursor cells (e.g ., HEK293T) cells using a standard transfection protocol (e.g., PEI).
  • precursor cells e.g ., HEK293T
  • PEI a standard transfection protocol
  • Viral particles are harvested from the media after a period of time (e.g., 72 h post transfection) and from the cells and media at a later point in time (e.g., 120 h post transfection).
  • Vims present in the media is concentrated by precipitation with 8% poly(ethylene glycol) and 500 mM sodium chloride and the precipitated vims is added to the lysates prepared from the collected cells.
  • Vimses are purified over iodixanol (Optiprep, Sigma) step gradients (15%, 25%, 40% and 60%). Vimses are concentrated and formulated in PBS. Vims titers are determined by measuring the number of DNasel-resistant vector genome copies (VGs) using qPCR and the linearized genome plasmid as a control.
  • VGs DNasel-resistant vector genome copies
  • the Rep protein can be selected from the group consisting of Rep78, Rep68, Rep52, and Rep40.
  • the genome of the AAV helper vims comprises an AAV helper gene selected from the group consisting of E2, E4, and VA.
  • the second nucleic acid and the first nucleic acid can be in trans.
  • the second nucleic acid and the first nucleic acid can be in cis.
  • the cell can be selected from a group consisting of a human, a primate, a murine, a feline, a canine, a porcine, an ovine, a bovine, an equine, an epine, a caprine and a lupine host cell.
  • the cell is a progenitor or precursor cell, such as a stem cell.
  • the stem cell is a mesenchymal cell, embryonic stem cell, induced pluripotent stem cell (iPSC), fibroblast or other tissue specific stem cell.
  • the cell can be immortalized.
  • the immortalized cell is a HEK293cell.
  • the cell is a differentiated cell. Based on the disclosure provided, it is expected that this system can be used in conjunction with any transgenic line expressing a recombinase in the target cell type of interest to develop AAV capsids that more efficiently transduce that target cell population.
  • a heterologous nucleic acid e.g ., therapeutic nucleic acid or transgene disclosed herein
  • the transgene may be encapsidated by a recombinant AAV (rAAV) capsid protein or rAAV particle such as those described herein.
  • rAAV recombinant AAV
  • Methods may be ex vivo , e.g., scientific research purposes or for producing adoptive cellular therapies.
  • the subject may be a human primary cell or a mature cell, or cell line.
  • the subject may be a cell from a monkey, hamster, or mouse. In either case, delivery may include contacting the composition with the cell or cell line.
  • Methods may be in vivo, e.g., treating a disease or a condition in a subject in need thereof.
  • the subject may be mammal, such as a human or non-human primate, in which case delivery of the composition may comprise administering the composition to the subject.
  • delivery of the heterologous nucleic acid comprises
  • methods of increasing transduction of a heterologous nucleic acid in a target in vivo environment comprise delivering a rAAV particle described herein, the rAAV engineered to have an increased transduction efficiency in a target in vivo environment (e.g., tissue or cell type).
  • a target in vivo environment e.g., tissue or cell type
  • the increased transduction efficiency comprises a 1- fold, 2-fold, 3-fold, 4-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, 11-fold, 12-fold, 13- fold, 14-fold, 15-fold, 16-fold, 17-fold, 18-fold, 19-fold, 20-fold, 21-fold, 22-fold, 23-fold, 24- fold, 25-fold, 26-fold, 27 -fold, 28-fold, 29-fold, 30-fold, 31-fold, 32-fold, 33-fold, 34-fold, 35- fold, 36-fold, 37-fold, 38-fold, 39-fold, 40-fold, 41-fold, 42-fold, 43-fold, 44-fold, 45-fold, 46- fold, 47-fold, 48-fold, 49-fold, 50-fold, 75-fold, or 100-fold increase, or more, relative to a reference AAV.
  • the increased transduction efficiency is at least 30-fold. In some instances, the increased transduction efficiency is at least 40-fold. In some instances, the increased transduction efficiency is at least 50-fold. In some instances, the increased transduction efficiency is at least 60-fold. In some instances, the increased transduction efficiency is at least 80-fold. In some instances, the increased transduction efficiency is at least 90-fold. In some instances, the increased transduction efficiency is at least 100-fold.
  • Methods of delivering a heterologous nucleic acid to a target in vivo environment comprising delivering the rAAV particle described herein that has been engineered to have an increased specificity in a target in vivo environment (e.g., tissue or cell type), as compared to a reference AAV.
  • a target in vivo environment e.g., tissue or cell type
  • Methods comprise detecting whether a rAAV possesses more specificity for a target in vivo environment than a reference AAV, includes measuring a level of gene expression product (e.g., RNA or protein) expressed from the heterologous nucleic acid encapsidated by the rAAV in a tissue sample obtained from the target in vivo environment in a subject; and comparing the measured level to a control level (such as, for e.g., the gene expression product expressed from a heterologous nucleic acid encapsidated by a reference AAV (e.g., AAV9)).
  • a control level such as, for e.g., the gene expression product expressed from a heterologous nucleic acid encapsidated by a reference AAV (e.g., AAV9).
  • a control level such as, for e.g., the gene expression product expressed from a heterologous nucleic acid encapsidated by a reference AAV (e.g.
  • the reference AAV has a serotype selected from the group consisting of AAV1, AAV2, AAV3, AAV4, AAV5, AAV6, AAV7, AAV8, AAV9, AAV10, AAV 11, AAV 12, or variant thereof.
  • the reference AAV can have a serotype selected from the group consisting of AAV-PHP.B, AAV-PHP.eB, and AAV-PHP.S.
  • CNS central nervous system
  • the rAAV particle encapsidating the heterologous nucleic acid comprises a rAAV capsid protein engineered with an increased specificity and, in some cases, transduction efficiency when measured in the CNS or the PNS of the subject, even when administered to the subject systemically, as compared to a reference AAV.
  • Methods comprise delivering a rAAV particle comprising an rAAV capsid protein with increased specificity and/or transduction efficiency when measured in the CNS in the subject, as compared to a reference AAV (e.g ., AAV9).
  • delivery is systemic.
  • delivery is direct (e.g., into the affected area of the CNS).
  • the rAAV capsid protein may comprise a substitution of at least one, two, three, four, five, six, seven, eight, nine, ten, or eleven amino acids provided in an amino acid sequence provided in any one of Tables 2-3, in an amino acid sequence of a parental AAV.
  • XI is selected from the group consisting of A, E, D, G, R, S and T.
  • the insertion further comprises two amino acids, wherein X2 is selected from the group consisting of A, G, I, L, M, N, Q, R, T, and Y.
  • the insertion further comprises three amino acids, wherein X3 is selected from the group consisting of E, K, L, T, and Q.
  • the insertion further comprises at least four amino acids, wherein XI is selected from the group consisting of A, E, D, G, R, S and T, X2 is selected from the group consisting of A, G, I, L, M, N, Q, R, T, and Y, X3 is selected from the group consisting of E, K, L, T, and Q, and X4 is selected from the group consisting of G, I, K, L, M, R, T, and V. In some instances, the insertion further comprises five amino acids wherein X5 is selected from the group consisting of A, D, G, P, L, Q, and V.
  • the insertion further comprises at least six amino acids, wherein X6 is selected from the group consisting of F, K, L, N, P, Q, S, and V. In some instances, the insertion further comprises at least seven amino acids, wherein X7 is selected from the group consisting of I, K, L, P, S, and V.
  • a rAAV particle encapsidating a heterologous nucleic acid to a CNS in a subject comprising (i) an increased specificity and/or transduction efficiency of the heterologous nucleic acid for the CNS, wherein the rAAV particle has an rAAV capsid protein comprising an insertion of at least or about three, four, five, six, or seven amino acids of an amino acid sequence TALKPFL, TTLKPFL,
  • the delivering is systemic.
  • the delivery is direct (e.g., injected into the in vivo environment).
  • the parental AAV capsid protein is AAV9 capsid protein (for e.g., provided in SEQ ID NO: 1).
  • delivery is more specific than a delivery of the heterologous nucleic acid by a reference AAV, e.g., AAV9.
  • the delivery is systemic (e.g., intravenous).
  • the subject is a mammal. In some embodiments, the subject is a human.
  • the methods of delivering a heterologous nucleic acid comprise delivering to a target in vivo environment in a subject a composition, the composition comprising a rAAV particle with a rAAV capsid protein, the rAAV capsid protein encapsidating a viral vector encoding a heterologous nucleic acid (e.g., therapeutic nucleic acid).
  • the target in vivo environment is the liver.
  • the rAAV particle encapsidating the heterologous nucleic acid comprises a rAAV capsid protein engineered with an increased specificity and, in some cases, transduction efficiency when measured in the target in vivo environment of the subject, even when administered to the subject systemically.
  • methods comprise delivering a rAAV particle comprising an rAAV capsid protein with increased specificity and/or transduction efficiency of the
  • rAAVs optimized for targeting the liver have amino acid sequences that comprise an amino acid sequence provided in SEQ ID NOS: 950-1031 and 15054-15146 (FIG. 35).
  • the rAAV capsid protein suitable for delivery of the heterologous nucleic acid to the liver can comprise an insertion of at least one amino acid in a parental AAV capsid protein.
  • the insertion comprises at least one, two, three, four, five, six, seven, eight, nine, ten, or eleven amino acids provided in an amino acid sequence provided in Table 4, or FIG. 35.
  • a rAAV particle encapsidating a heterologous nucleic acid to the target in vivo environment selected from the group consisting of the liver in a subject, the rAAV particle comprising an increased specificity and/or transduction efficiency of the heterologous nucleic acid for the target in vivo environment, wherein the rAAV particle has an rAAV capsid protein comprising an insertion of at least or about three, four, five, six, seven, eight, nine, ten, or eleven amino acids of an amino acid sequence provided in Table 4, or FIG. 35.
  • delivery is more specific than a delivery of the
  • methods further comprise reducing or ablating delivery of the heterologous nucleic acid in an off-target in vivo environment, such as the liver, compared to a reference AAV.
  • delivery is characterized by an increase in efficiency of transduction (e.g., of the heterologous nucleic acid) in the target in vivo environment than a transduction efficiency in the target in vivo environment of the reference AAV.
  • the delivery is systemic (e.g., intravenous).
  • the subject is a mammal. In some cases, the mammal is a human.
  • compositions e.g., rAAV particle, AAV vector, pharmaceutical composition
  • the composition is a rAAV capsid protein described herein.
  • the composition is an isolated and purified rAAV capsid protein described herein.
  • the rAAV particle encapsidates an AAV vector comprising a transgene (e.g., therapeutic nucleic acid).
  • the composition is a rAAV capsid protein described herein conjugated with a therapeutic agent disclosed herein.
  • the composition is a pharmaceutical composition comprising the rAAV particle and a pharmaceutically acceptable carrier.
  • the one or more compositions are administered to the subject alone (e.g., standalone therapy).
  • the one or more compositions are administered in combination with an additional agent.
  • the composition is a first-line therapy for the disease or condition.
  • the composition is a second-line, third-line, or fourth-line therapy, for the disease or condition.
  • kits for treating a disease or a condition, or a symptom of the disease or condition, in a subject comprising: (a) diagnosing a subject with a disease or a condition affecting a target in vivo environment; and (b) treating the disease or the condition by administering to the subject a therapeutically effective amount of a composition disclosed herein (e.g ., rAAV particle, AAV vector, pharmaceutical composition), wherein the composition is engineered with an increased specificity for the target in vivo environment.
  • a composition disclosed herein e.g ., rAAV particle, AAV vector, pharmaceutical composition
  • Disclosed herein are methods of treating a disease or a condition, or a symptom of the disease or the condition, afflicting a target in vivo environment in a subject comprising: (a) administering to the subject a composition (e.g., rAAV particle, AAV vector, pharmaceutical composition); and (b) expressing the therapeutic nucleic acid into a target in vivo environment in the subject with an increased specificity and/or transduction efficiency, as compared to a reference AAV.
  • the reference AAV is AAV9, or a variant thereof.
  • Methods of treating a disease or condition affecting the central nervous system comprise administering a rAAV particle to a CNS in a subject, the rAAV particle comprising an rAAV capsid protein comprising an insertion of at least or about three, four, five, six, seven, eight, nine, ten, or eleven amino acids of an amino acid sequence TALKPFL, TTLKPFL, TLQIPFK, TMQKPFI, RYQGDSV, or TTLKPFS, or any amino acid sequence provided in
  • the parental AAV capsid protein is AAV9 capsid protein (for e.g., provided in SEQ ID NO: 1.
  • the parental AAV capsid protein comprises an amino acid sequence that is at least 95%, 96%, 96.1, 96.2%, 96.3%, 96.4%, 96.5%, 96.6%, 96.7%, 96.8%, 96.9%, 97.0%, 97.1%, 97.2%, 97.3%, 97.4%,
  • delivery is more specific than a delivery of the heterologous nucleic acid by a reference AAV, e.g., AAV9.
  • the delivery is systemic (e.g., intravenous).
  • the subject is a human or a non-human primate.
  • Methods of treating a disease or a condition afflicting a target in vivo environment comprising a liver comprise administering a rAAV particle to the target in vivo environment in a subject, the rAAV particle comprising an rAAV capsid protein comprising a substitution of at least or about three, four, five, six, seven, eight, nine, ten, or eleven, amino acids of an amino acid sequence provided in any one of SEQ ID NOS: 950-1031 and 15054-15146 (FIG. 35).
  • methods comprise delivering a rAAV particle comprising an rAAV capsid protein with increased specificity for the liver in the subject, as compared to a reference AAV (e.g., AAV9).
  • rAAVs optimized for targeting the liver have amino acid sequences comprising an amino acid sequence KAYSVQV, PSGSARS, and RTANALG at an amino acid position 588_589 in a parental AAV capsid protein.
  • the parental AAV capsid protein is AAV9 capsid protein (for e.g., provided in SEQ ID NO: 1).
  • the parental AAV capsid protein comprises an amino acid sequence that is at least 95%, 96%, 96.1, 96.2%, 96.3%, 96.4%, 96.5%, 96.6%, 96.7%, 96.8%, 96.9%, 97.0%, 97.1%, 97.2%, 97.3%, 97.4%, 97.5%, 97.6%, 97.7%, 97.8%, 97.9%, 98.0%, 98.1%, 98.2%, 98.3%, 98.4%, 98.5%, 98.6%, 98.7%, 98.7%, 98.8%, 98.9%, 99.0%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, or 100.0% identical to SEQ ID NO: 1.
  • delivery is more specific than a delivery of the heterologous nucleic acid by a reference AAV, e.g., AAV9.
  • the delivery is systemic (e.g., intravenous or intranasal).
  • the subject is a human or a non-human primate.
  • methods of modulating a target gene expression product comprising administering to a subject in need thereof a composition (e.g., rAAV particle, AAV vector, pharmaceutical composition) disclosed herein.
  • a composition e.g., rAAV particle, AAV vector, pharmaceutical composition
  • methods provided herein comprise administering to a subject a rAAV with a rAAV capsid protein encapsidating a viral vector comprising a heterologous nucleic acid that modulates the expression or the activity of the target gene expression product.
  • the disease or the condition is characterized by an increased or enhanced expression or activity of a gene or gene expression product thereof, as compared to a normal individual.
  • administering the therapeutically effective amount of the composition restores the expression or the activity of the gene or gene expression product thereof to a level that is typical in a normal individual.
  • normal individual refers to an individual that is not afflicted with the disease or the condition characterized by the variation in expression or activity of the gene or gene expression product thereof.
  • Non-limiting examples of genes involved in central nervous system (CNS) diseases or disorders include Sarcoglycan Alpha (SGCA), glutamic acid decarboxylase 65 (GAD65), glutamic acid decarboxylase 67 (GAD67), CLN2 gene, Nerve Growth Factor (NGF), glial cell derived neurotrophic factor (GDNF), Neurturin, Survival Of Motor Neuron 1, Telomeric (SMN1), b-Glucocerebrosidase (GCase), Frataxin (FXN), Huntingtin (HTN), methyl-CpG binding protein 2 (MECP2), peroxisomal biogenesis factor (PEX), progranulin (GRN), an antitubulin agent, copper-zinc superoxide dismutase (SOD1), Glucosylceramidase Beta (GBA), NPC Intracellular Cholesterol Transporter 1 (NPC1), and NPS3.
  • SGCA Sarcoglycan Alpha
  • GAD65 glutamic acid decar
  • the peroxisomal biogenesis factor is selected from the group consisting of PEX 1, PEX2, PEX3, PEX4, PEX5, PEX6, PEX7, PEX10, RECI Ib, PEX 12, PEX13, PEX14, PEX 16, PEX 19, and PEX26.
  • Non-limiting examples of genes implicated in disease or disorder of a particular organ include Cystic Fibrosis Transmembrane Conductance Regulator (CFTR), Factor X (FIX), RPE65, Retinoid Isomerohydrolase (RPE65), Sarcoglycan Alpha (SGCA), and sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a).
  • CFTR Cystic Fibrosis Transmembrane Conductance Regulator
  • FIX Factor X
  • RPE65 Retinoid Isomerohydrolase
  • SGCA Sarcoglycan Alpha
  • SERCA2a sarco/endoplasmic reticulum Ca2+-ATPase
  • the composition is administered at dosage levels sufficient to deliver from about 0.0001 mg/kg to about 100 mg/kg, from about 0.001 mg/kg to about 0.05 mg/kg, from about 0.005 mg/kg to about 0.05 mg/kg, from about 0.001 mg/kg to about 0.005 mg/kg, from about 0.05 mg/kg to about 0.5 mg/kg, from about 0.01 mg/kg to about 50 mg/kg, from about 0.1 mg/kg to about 40 mg/kg, from about 0.5 mg/kg to about 30 mg/kg, from about 0.01 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, or from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
  • the viral genome (vg) concentration of the composition that is administered is between 1.0 xlO 11 vg per kilogram (kg) and 1.0 xl0 16 vg/kg.
  • the concentration of infectious particles of at least or about 10 7 , 10 8 , 10 9 , 10 10 , 10 11 , 10 12 , 10 13 , 10 14 , 10 15 , 10 16 , or 10 17 .
  • the concentration of infectious particles is 2xl0 7 , 2xl0 8 , 2xl0 9 , 2xl0 10 , 2xlO n , 2xl0 12 , 2xl0 13 , 2xl0 14 , 2xl0 15 , 2xl0 16 , or 2xl0 17 .
  • the concentration of infectious particles is 2xl0 7 , 2xl0 8 , 2xl0 9 , 2xl0 10 , 2xlO n , 2xl0 12 , 2xl0 13 , 2xl0 14 , 2xl0 15 , 2xl0 16 , or 2xl0 17 .
  • the concentration of infectious particles is 2xl0 7 , 2xl0 8 , 2xl0 9 , 2xl0 10 , 2xlO n , 2xl0 12 , 2xl0 13 , 2xl0 14 , 2xl0 15 , 2xl0 16 , or 2x
  • concentration of the infectious particles 3xl0 7 , 3xl0 8 , 3xl0 9 , 3xl0 10 , 3xl0 n , 3xl0 12 , 3xl0 13 , 3xl0 14 , 3xl0 15 , 3xl0 16 , or 3xl0 17 .
  • concentration of the infectious particles 4xl0 7 , 4xl0 8 , 4xl0 9 , 4xl0 10 , 4xlO n , 4xl0 12 , 4xl0 13 , 4xl0 14 , 4xl0 15 , 4xl0 16 , or 4xl0 17 the concentration of the infectious particles 4xl0 7 , 4xl0 8 , 4xl0 9 , 4xl0 10 , 4xlO n , 4xl0 12 , 4xl0 13 , 4xl0 14 , 4xl0 15 , 4xl0 16 , or 4xl0 17 .
  • the administering of step is performed once. Alternatively, the administering of step is repeated at least twice.
  • the administering of step may be performed once daily.
  • the administering of step comprises intravenous administration.
  • the administering comprises pulmonary administration.
  • the administering comprises intranasal administration (such as a spray).
  • the administering of step comprises injecting the composition into a target in vivo environment. In some cases, the administering of step does not comprise injecting the composition into the target in vivo environment.
  • Disclosed herein methods of delivering at least one of an AAV particle and viral vector to a subject for example - to treat or prevent a disease or condition in a subject.
  • the subject in some cases, is a mammal.
  • a mammal include a mouse, rat, guinea pig, rabbit, chimpanzee, or farm animal.
  • the mammal is a non-human primate.
  • the subject is human.
  • the subject of the present disclosure may not be diagnosed with a disease or condition.
  • the subject may be a patient that is diagnosed with a disease or disorder, or suspected of having the disease or the disorder.
  • rAAV spinal muscular atrophy
  • ALS amyotrophic lateral sclerosis
  • Parkinson's disease Pompe disease
  • Huntington's disease Alzheimer's disease
  • Battens disease lysosomal storage disorders
  • glioblastoma multiforme glioblastoma multiforme
  • Rett syndrome Leber's congenital amaurosis
  • Late infantile neuronal ceroid lipofuscinosis LINCL
  • the disease or the condition may, in some embodiments, be characterized by a reduced or ablated expression or activity of a gene or gene expression product thereof, as compared to a normal individual. In some embodiments, be characterized by an increased or enhanced expression or activity of a gene or gene expression product thereof, as compared to a normal individual.
  • the disease or condition is localized to a particular in vivo environment in the subject, e.g., the brain or the liver.
  • the compositions of the present disclosure are particularly useful for the treatment of the diseases or conditions described herein because they specifically target the in vivo environment and deliver a therapeutic nucleic acid engineered to modulate the activity or the expression of a target gene expression product involved with the pathogenesis or pathology of the disease or condition.
  • the disease or condition comprises a disease or condition of the central nervous system (CNS).
  • CNS central nervous system
  • Non-limiting examples of disease of the CNS include Absence of the Septum Pellucidum, Acid Lipase Disease, Acid Maltase Deficiency, Acquired Epileptiform Aphasia, Acute Disseminated Encephalomyelitis, Attention Deficit-Hyperactivity Disorder (ADHD), Adie's Pupil, Adie's Syndrome, Adrenoleukodystrophy, Agenesis of the Corpus Callosum, Agnosia, Aicardi Syndrome, Aicardi-Goutieres Syndrome Disorder, AIDS - Neurological Complications, Alexander Disease, Alpers' Disease, Alternating Hemiplegia, Alzheimer's Disease, Amyotrophic Lateral Sclerosis (ALS), Anencephaly, Aneurysm, Angelman Syndrome, Angiomatosis, Anoxia, Antiphospholipid Syndrome, Aphasia, Apraxia, Arachnoid Cysts,
  • Atrial Fibrillation and Stroke Attention Deficit-Hyperactivity Disorder
  • Autism Spectrum Disorder Autism Spectrum Disorder, Autonomic Dysfunction, Back Pain, Barth Syndrome, Batten Disease, Becker's Myotonia, Behcet's Disease, Bell's Palsy, Benign Essential Blepharospasm, Benign Focal Amyotrophy, Benign Intracranial Hypertension, Bernhardt-Roth Syndrome, Binswanger's Disease, Blepharospasm, Bloch-Sulzberger Syndrome, Brachial Plexus Birth Injuries, Brachial Plexus Injuries, Bradbury-Eggleston Syndrome, Brain and Spinal Tumors, Brain Aneurysm, Brain Injury, Brown-Sequard Syndrome, Bulbospinal Muscular Atrophy, Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL), Canavan Disease, Carpal Tunnel Syndrome, Causalgia, Cavernomas,
  • Choreoacanthocytosis Chronic Inflammatory Demyelinating Polyneuropathy (CIDP), Chronic Orthostatic Intolerance, Chronic Pain, Cockayne Syndrome Type II, Coffin Fowry Syndrome, Colpocephaly, Coma, Complex Regional Pain Syndrome, Congenital Facial Diplegia,
  • Encephalopathy familial infantile
  • Encephalotrigeminal Angiomatosis Epilepsy
  • Epileptic Hemiplegia Erb's Palsy, Erb-Duchenne and Dejerine-Klumpke Palsies
  • Essential Tremor Extrapontine Myelinolysis
  • Fabry Disease Fahr's Syndrome
  • Fainting Familial Dysautonomia
  • Familial Hemangioma Familial Idiopathic Basal Ganglia Calcification
  • Familial Periodic Paralyses Familial Spastic Paralysis, Farber's Disease, Febrile Seizures
  • Fipodystrophy Intracranial Cysts, Intracranial Hypertension, Isaacs' Syndrome, Joubert Syndrome, Kearns-Sayre Syndrome, Kennedy's Disease, Kinsbourne syndrome, Kleine-Fevin Syndrome, Klippel-Feil Syndrome, Klippel-Trenaunay Syndrome (KTS), Kliiver-Bucy
  • Mucopolysaccharidoses Multi-Infarct Dementia, Multifocal Motor Neuropathy, Multiple Sclerosis, Multiple System Atrophy, Multiple System Atrophy with Orthostatic Hypotension, Muscular Dystrophy, Myasthenia -Congenital, Myasthenia Gravis, Myelinoclastic Diffuse Sclerosis, Myoclonic Encephalopathy of Infants, Myoclonus, Myopathy, Myopathy- Congenital, Myopathy -Thyrotoxic, Myotonia, Myotonia Congenita, Narcolepsy, Neuroacanthocytosis, Neurodegeneration with Brain Iron Accumulation, Neurofibromatosis, Neuroleptic Malignant Syndrome, Neurological Complications of AIDS, Neurological Complications of Lyme Disease, Neurological Consequences of Cytomegalovirus Infection, Neurological Manifestations of Pompe Disease, Neurological Sequelae Of Lupus, Neuromyelitis Optica, Neuromyotonia, Neuronal Ceroid Lipofuscinos
  • Leukoencephalopathy Progressive Sclerosing Poliodystrophy, Progressive Supranuclear Palsy, Prosopagnosia, Pseudo-Torch syndrome, Pseudotoxoplasmosis syndrome, Pseudotumor Cerebri, Psychogenic Movement, Ramsay Hunt Syndrome I, Ramsay Hunt Syndrome II, Rasmussen's Encephalitis, Reflex Sympathetic Dystrophy Syndrome, Refsum Disease, Refsum Disease - Infantile, Repetitive Motion Disorders, Repetitive Stress Injuries, Restless Legs Syndrome, Retrovirus-Associated Myelopathy, Rett Syndrome, Reye's Syndrome, Rheumatic Encephalitis, Riley-Day Syndrome, Sacral Nerve Root Cysts, Saint Vitus Dance, Salivary Gland Disease, Sandhoff Disease, Schilder's Disease, Schizencephaly, Seitelberger Disease, Seizure Disorder, Semantic Dementia, Septo-Optic Dysplasia, Se
  • Panencephalitis Subcortical Arteriosclerotic Encephalopathy, Short-lasting, Unilateral, Neuralgiform (SUNCT) Headache, Swallowing Disorders, Sydenham Chorea, Syncope, Syphilitic Spinal Sclerosis, Syringohydromyelia, Syringomyelia, Systemic Lupus
  • VHL Von Economo's Disease
  • VHL Von Hippel-Lindau Disease
  • VHL Von Recklinghausen's Disease
  • Wallenberg's Syndrome Werdnig-Hoffman Disease
  • Wemicke- Korsakoff Syndrome West Syndrome
  • Whiplash Whipple's Disease
  • Williams Syndrome Wilson Disease
  • Wolman's Disease and X-Linked Spinal and Bulbar Muscular Atrophy.
  • the disease or condition comprises a liver disease or disorder, or is associated with a liver disease or disorder.
  • disorders of bile acid synthesis e.g ., Wilson disease, Progressive familial intrahepatic cholestasis type 3
  • disorders of carbohydrate metabolism e.g., Hereditary fructose intolerance, Glycogen storage disease type IV
  • disorders of amino acids metabolism e.g., tyrosinemia type I
  • Urea cycle disorders e.g., argininosuccinate lyase deficiency, citrin deficiency (CTLN2, NICCD)
  • disorders of lipid metabolism e.g., cholesteryl ester storage disease
  • others including but not limited to Alpha- 1 antitrypsin deficiency, cystic fibrosis, hereditary hemochromatosis, Alstrom syndrome, and congenital hepatic fibrosis.
  • the disease or condition is a disease or condition is of the liver.
  • liver diseases or disorders include Alagille Syndrome, Alcohol- Related Liver Disease, Alpha- 1 Antitrypsin Deficiency, Autoimmune Hepatitis, Benign Liver Tumors, Biliary Atresia, Cirrhosis, Crigler-Najjar Syndrome, Galactosemia, Gilbert Syndrome, Hemochromatosis, Hepatic Encephalopathy, Hepatitis A, Hepatitis B, Hepatitis C, Hepatorenal Syndrome, Intrahepatic Cholestasis of Pregnancy (ICP), Lysosomal Acid Lipase Deficiency (LAL-D), Liver Cysts, Liver Cancer, Newborn Jaundice, Non-Alcoholic Fatty Liver Disease, Primary Biliary Cholangitis (PBC), Primary Sclerosing Cholangitis (PSC), Reye Syndrome, Type I Glycogen Storage Disease,
  • ICP Pregnancy
  • kits for treating a disease or a condition associated with an aberrant expression or activity of a target gene or gene expression product thereof comprising modulating the expression or the activity of a target gene or gene expression product in a subject by administering a rAAV encapsidating a heterologous nucleic acid of the present disclosure.
  • administration is systemic administration.
  • the expression or the activity of the target gene or gene expression product is decreased, relative to that in a normal (non-diseased) individual; and administering the rAAV to the subject is sufficient to increase the expression of the activity of the target gene or gene expression product to that of a normal individual.
  • the expression or the activity of the gene or gene expression product is increased, relative to that in a normal individual; and administering the rAAV to the subject is sufficient to decrease the expression or the activity of the target gene or gene expression product.
  • a subject diagnosed with Alzheimer’s disease which is caused, in some cases, by a gain-of-function of a Presenilin 1 and/or Presenilin 2 (encoded by the gene PSEN 1 and PSEN2, respectively) is administered a rAAV disclosed herein encapsidating a therapeutic nucleic acid that is a silencing RNA (siRNA), or other RNAi with a loss-of-function effect on PSEN 1 mRNA.
  • siRNA silencing RNA
  • Also provided are methods of treating or preventing a disease or condition disclosed herein in a subject comprising administering to the subject a therapeutically effective amount of an AAV vector comprising a nucleic acid sequence encoding a therapeutic gene expression product described herein.
  • the AAV vector may be encapsidated in the modified capsid protein or AAV viral particle described herein.
  • the therapeutic gene expression product is effective to modulate the activity or expression of a target gene or gene expression product.
  • methods disclosed herein comprise administering a therapeutic rAAV composition by systemic administration. In some instances, methods comprise administering a therapeutic rAAV composition by oral administration. In some instances, methods comprise administering a therapeutic rAAV composition by intraperitoneal injection. In some instances, methods comprise administering a therapeutic rAAV composition in the form of an anal suppository. In some instances, methods comprise administering a therapeutic rAAV
  • compositions by intravenous (“i.v.”) administration may also administer therapeutic rAAV compositions disclosed herein by other routes, such as
  • methods comprise administering a therapeutic rAAV composition by topical administration, example, by brushing or otherwise contacting the rAAV composition to a region of the subject (e.g., eardrum, bladder).
  • routes for local delivery closer to site of injury or inflammation are preferred over systemic routes. Routes, dosage, time points, and duration of administrating therapeutics may be adjusted.
  • administration of therapeutics is prior to, or after, onset of either, or both, acute and chronic symptoms of the disease or condition.
  • An effective dose and dosage of pharmaceutical compositions to prevent or treat the disease or condition disclosed herein is defined by an observed beneficial response related to the disease or condition, or symptom of the disease or condition.
  • Beneficial response comprises preventing, alleviating, arresting, or curing the disease or condition, or symptom of the disease or condition.
  • the beneficial response may be measured by detecting a measurable improvement in the presence, level, or activity, of biomarkers, transcriptomic risk profile, or intestinal microbiome in the subject.
  • An“improvement,” as used herein refers to shift in the presence, level, or activity towards a presence, level, or activity, observed in normal individuals ( e.g . individuals who do not suffer from the disease or condition).
  • the dosage amount and/or route of administration may be changed, or an additional agent may be administered to the subject, along with the therapeutic rAAV composition.
  • the patient as a patient is started on a regimen of a therapeutic rAAV composition, the patient is also weaned off (e.g., step-wise decrease in dose) a second treatment regimen.
  • compositions in accordance with the present disclosure may be administered at dosage levels sufficient to deliver from about 0.0001 mg/kg to about 100 mg/kg, from about 0.001 mg/kg to about 0.05 mg/kg, from about 0.005 mg/kg to about 0.05 mg/kg, from about 0.001 mg/kg to about 0.005 mg/kg, from about 0.05 mg/kg to about 0.5 mg/kg, from about 0.01 mg/kg to about 50 mg/kg, from about 0.1 mg/kg to about 40 mg/kg, from about 0.5 mg/kg to about 30 mg/kg, from about 0.01 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, or from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic, diagnostic, or prophylactic, effect. It will be understood that the above dosing concentrations may be converted to vg or viral genomes per kg or into total viral genomes administered by one
  • a dose of the pharmaceutical composition may comprise a
  • concentration of infectious particles of at least or about 10 7 , 10 8 , 10 9 , 10 10 , 10 11 , 10 12 , 10 13 , 10 14 , 10 15 , 10 16 , or 10 17 .
  • the concentration of infectious particles is 2xl0 7 , 2xl0 8 , 2xl0 9 , 2xl0 10 , 2xlO n , 2xl0 12 , 2xl0 13 , 2xl0 14 , 2xl0 15 , 2xl0 16 , or 2xl0 17 .
  • the concentration of infectious particles is 2xl0 7 , 2xl0 8 , 2xl0 9 , 2xl0 10 , 2xlO n , 2xl0 12 , 2xl0 13 , 2xl0 14 , 2xl0 15 , 2xl0 16 , or 2xl0 17 .
  • the concentration of infectious particles is 2xl0 7 , 2xl0 8 , 2xl0
  • concentration of the infectious particles is 3xl0 7 , 3xl0 8 , 3xl0 9 , 3xl0 10 , 3xl0 u , 3xl0 12 , 3xl0 13 , 3xl0 14 , 3xl0 15 , 3xl0 16 , or 3xl0 17 .
  • the concentration of the infectious particles is 4xl0 7 , 4xl0 8 , 4xl0 9 , 4xl0 10 , 4xlO n , 4xl0 12 , 4xl0 13 , 4xl0 14 , 4xl0 15 , 4xl0 16 , or 4xl0 17 .
  • the concentration of the infectious particles is 5xl0 7 , 5xl0 8 , 5xl0 9 , 5xl0 10 , 5xl0 n , 5xl0 12 , 5xl0 13 , 5xl0 14 , 5xl0 15 , 5xl0 16 , or 5xl0 17 .
  • the concentration of the infectious particles is 6xl0 7 , 6xl0 8 , 6xl0 9 , 6xl0 10 , 6xlO u , 6xl0 12 , 6xl0 13 , 6xl0 14 , 6xl0 15 , 6xl0 16 , or 6x10 .
  • the concentration of the infectious particles is 7x10 , 7x10 , 7x10 , 7x10 , 7xl0 10 , 7xlO n , 7xl0 12 , 7xl0 13 , 7xl0 14 , 7xl0 15 , 7xl0 16 , or 7xl0 17 .
  • the concentration of the infectious particles is 8xl0 7 , 8x10 s , 8xl0 9 , 8xl0 10 , 8xl0 u , 8xl0 12 , 8xl0 13 , 8xl0 14 , 8xl0 15 , 8xl0 16 , or 8xl0 17 .
  • the concentration of the infectious particles is 9xl0 7 , 9xl0 8 , 9xl0 9 , 9xl0 10 , 9xlO n , 9xl0 12 , 9xl0 13 , 9xl0 14 , 9xl0 15 , 9xl0 16 , or 9xl0 17 .
  • compositions described herein suitable for delivery of the rAAV compositions described herein, as well as suitable dosing and treatment regimens for using the particular compositions described herein in a variety of treatment regimens.
  • the amount of therapeutic gene expression product in each therapeutically-useful composition may be prepared is such a way that a suitable dosage will be obtained in any given unit dose of the compound.
  • Factors such as solubility, bioavailability, biological half-life, route of
  • the rAAV compositions are suitably formulated pharmaceutical compositions disclosed herein, to be delivered either intraocularly, intravitreally, parenterally, subcutaneously, intravenously, intracerebro-ventricularly, intramuscularly, intrathecally, orally, intraperitoneally, by oral or nasal inhalation, or by direct injection to one or more cells, tissues, or organs by direct injection.
  • compositions suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g ., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils.
  • Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens,
  • chlorobutanol phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • the solution may be suitably buffered, if necessary, and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • these particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration. Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject.
  • preparations should meet sterility, pyrogenicity, and the general safety and purity standards as required by FDA Office of Biologies standards.
  • sterile injectable solutions comprising the rAAV compositions disclosed herein, which are prepared by incorporating the rAAV compositions disclosed herein in the required amount in the appropriate solvent with several of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile- filtered solution thereof.
  • Injectable solutions may be advantageous for systemic administration, for example by intravenous administration.
  • compositions in a neutral or salt form include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. Upon formulation, solutions will be
  • the formulations are easily administered in a variety of dosage forms such as injectable solutions, drug-release capsules, and the like.
  • formulations may comprise dry particles comprising active ingredients.
  • dry particles may have a diameter in the range from about 0.5 nm to about 7 nm or from about 1 nm to about 6 nm.
  • formulations may be in the form of dry powders for administration using devices comprising dry powder reservoirs to which streams of propellant may be directed to disperse such powder.
  • self-propelling solvent/powder dispensing containers may be used.
  • active ingredients may be dissolved and/or suspended in low-boiling propellant in sealed containers.
  • Such powders may comprise particles wherein at least 98% of the particles by weight have diameters greater than 0.5 nm and at least 95% of the particles by number have diameters less than 7 nm. Alternatively, at least 95% of the particles by weight have a diameter greater than 1 nm and at least 90% of the particles by number have a diameter less than 6 nm.
  • Dry powder compositions may include a solid fine powder diluent such as sugar and are conveniently provided in a unit dose form.
  • Low boiling propellants generally include liquid propellants having a boiling point of below 65 °F at atmospheric pressure. Generally, propellants may constitute 50% to 99.9% (w/w) of the composition, and active ingredient may constitute 0.1% to 20% (w/w) of the composition.
  • Propellants may further comprise additional ingredients such as liquid non-ionic and/or solid anionic surfactant and/or solid diluent (which may have particle sizes of the same order as particles comprising active ingredients).
  • compositions formulated for pulmonary delivery may provide active ingredients in the form of droplets of solution and/or suspension.
  • Such formulations may be prepared, packaged, and/or sold as aqueous and/or dilute alcoholic solutions and/or suspensions, optionally sterile, comprising active ingredients, and may conveniently be administered using any nebulization and/or atomization device.
  • Such formulations may further comprise one or more additional ingredients including, but not limited to, a flavoring agent such as saccharin sodium, a volatile oil, a buffering agent, a surface-active agent, and/or a preservative such as methylhydroxybenzoate.
  • Droplets provided by this route of administration may have an average diameter in the range from about 0.1 nm to about 200 nm.
  • Formulations described herein useful for pulmonary delivery may also be useful for intranasal delivery.
  • formulations for intranasal administration comprise a coarse powder comprising the active ingredient and having an average particle size from about 0.2 pm to 500 pm. Such formulations are administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close to the nose.
  • Formulations suitable for nasal administration may, for example, comprise from about as little as 0.1% (w/w) and as much as 100% (w/w) of active ingredient, and may comprise one or more of the additional ingredients described herein.
  • a pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for buccal administration. Such formulations may, for example, be in the form of tablets and/or lozenges made using
  • formulations suitable for buccal administration may comprise powders and/or an aerosolized and/or atomized solutions and/or suspensions comprising active ingredients.
  • Such powdered, aerosolized, and/or aerosolized formulations, when dispersed, may comprise average particle and/or droplet sizes in the range of from about 0.1 nm to about 200 nm, and may further comprise one or more of any additional ingredients described herein.
  • Suitable dose and dosage administrated to a subject is determined by factors including, but not limited to, the particular therapeutic rAAV composition, disease condition and its severity, the identity (e.g ., weight, sex, age) of the subject in need of treatment, and can be determined according to the particular circumstances surrounding the case, including, e.g., the specific agent being administered, the route of administration, the condition being treated, and the subject or host being treated.
  • AAV compositions The amount of AAV compositions and time of administration of such compositions will be within the purview of the skilled artisan having benefit of the present teachings. It is likely, however, that the administration of therapeutically-effective amounts of the disclosed compositions may be achieved by a single administration, example, a single injection of sufficient numbers of infectious particles to provide therapeutic benefit to the patient undergoing such treatment. This is made possible, at least in part, by the fact that certain target cells (e.g neurons) do not divide, obviating the need for multiple or chronic dosing.
  • target cells e.g neurons
  • the number of infectious particles administered to a mammal may be on the order of about 10 7 , 10 8 , 10 9 , 10 10 , 10 11 , 10 12 , 10 13 , or even higher, infectious particles/ml given either as a single dose, or divided into two or more administrations as may be required to achieve therapy of the particular disease or disorder being treated.
  • the daily and unit dosages are altered depending on a number of variables including, but not limited to, the activity of the therapeutic rAAV composition used, the disease or condition to be treated, the mode of administration, the requirements of the individual subject, the severity of the disease or condition being treated, and the judgment of the practitioner.
  • the administration of the therapeutic rAAV composition is hourly, once every 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, 18 hours, 19 hours, 20 hours, 21 hours 22 hours, 23 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days,
  • the effective dosage ranges may be adjusted based on subject’s response to the treatment. Some routes of administration will require higher concentrations of effective amount of therapeutics than other routes.
  • the administration of therapeutic rAAV composition is administered chronically, that is, for an extended period of time, including throughout the duration of the patient’s life in order to ameliorate or otherwise control or limit the symptoms of the patient’s disease or condition.
  • the dose of therapeutic rAAV composition being administered may be temporarily reduced or temporarily suspended for a certain length of time (i.e., a“drug holiday”).
  • the length of the drug holiday is between 2 days and 1 year, including by way of example only, 2 days, 3 days, 4 days,
  • the dose reduction during a drug holiday is, by way of example only, by 10%-100%, including by way of example only 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, and 100%.
  • the dose of drug being administered may be temporarily reduced or temporarily suspended for a certain length of time (i.e., a“drug diversion”).
  • the length of the drug diversion is between 2 days and 1 year, including by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, or more than 28 days.
  • the dose reduction during a drug diversion is, by way of example only, by 10% -100%, including by way of example only 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%,
  • the normal dosing schedule is optionally reinstated.
  • a maintenance dose is administered if necessary. Subsequently, in specific embodiments, the dosage or the frequency of administration, or both, is reduced, as a function of the symptoms, to a level at which the improved disease, disorder or condition is retained. In certain embodiments, however, the patient requires intermittent treatment on a long-term basis upon any recurrence of symptoms.
  • Toxicity and therapeutic efficacy of such therapeutic regimens are determined by standard pharmaceutical procedures in cell cultures or experimental animals, including, but not limited to, the determination of the LD50 and the ED50.
  • the dose ratio between the toxic and therapeutic effects is the therapeutic index and it is expressed as the ratio between LD50 and ED50.
  • the data obtained from cell culture assays and animal studies are used in formulating the therapeutically effective daily dosage range and/or the therapeutically effective unit dosage amount for use in mammals, including humans.
  • the dosage amount of the therapeutic rAAV composition described herein lies within a range of circulating concentrations that include the ED50 with minimal toxicity.
  • the daily dosage range and/or the unit dosage amount varies within this range depending upon the dosage form employed and the route of administration utilized.
  • a therapeutic nucleic acid may be used alone or in combination with an additional therapeutic agent (together,“therapeutic agents”).
  • an“additional therapeutic agent” as used herein is administered alone.
  • the therapeutic agent may be administered together or sequentially in a combination therapy.
  • the combination therapy may be administered within the same day, or may be administered one or more days, weeks, months, or years apart.
  • a therapeutic nucleic acid provided herein is administered if the subject is determined to be non-responsive to a first line of therapy.
  • the additional therapeutic agent can comprise a small molecule.
  • the additional therapeutic agent can comprise an antibody, or antigen-binding fragment.
  • the additional therapeutic agent can comprise a cell-based therapy.
  • Exemplary cell-based therapies include without limitation immune effector cell therapy, chimeric antigen receptor T-cell (CAR-T) therapy, natural killer cell therapy and chimeric antigen receptor natural killer (NK) cell therapy.
  • CAR-T chimeric antigen receptor T-cell
  • NK chimeric antigen receptor natural killer
  • Either NK cells, or CAR-NK cells, or a combination of both NK cells and CAR-NK cells can be used in combination with the methods disclosed herein.
  • the NK cells and CAR-NK cells are derived from human induced pluripotent stem cells (iPSC), umbilical cord blood, or a cell line.
  • the NK cells and CAR-NK cells can comprise a cytokine receptor and a suicide gene.
  • the cell-based therapy can comprises a stem cell therapy.
  • the stem cells may be expanded adipose- derived stem cells (eASCs), hematopoietic stem cells (HSCs), mesenchymal stem (stromal) cells (MSCs), or induced pluripotent stem cells (iPSCs) derived from the cells of the subject.
  • eASCs expanded adipose- derived stem cells
  • HSCs hematopoietic stem cells
  • MSCs mesenchymal stem
  • iPSCs induced pluripotent stem cells
  • kits comprising compositions disclosed herein. Also disclosed herein are kits for the treatment or prevention of a disease or conditions of the central nervous system (CNS), or target organ or environment (e.g., liver).
  • the disease or condition is cancer, a pathogen infection, pulmonary disease or condition, neurological disease, muscular disease, or an immune disorder, such as those described herein.
  • a kit can include a therapeutic or prophylactic composition containing an effective amount of a composition of a rAAV particle encapsidating a recombinant AAV vector encoding a therapeutic nucleic acid (e.g., therapeutic nucleic acid) and a recombinant AAV (rAAV) capsid protein of the present disclosure.
  • a kit can include a therapeutic or prophylactic composition containing an effective amount of cells modified by the rAAV described herein (“modified cell”), in unit dosage form that express therapeutic nucleic acid.
  • modified cell modified cell
  • a kit comprises a sterile container which can contain a therapeutic composition; such containers can be boxes, ampules, bottles, vials, tubes, bags, pouches, blister-packs, or other suitable container forms known in the art. Such containers can be made of plastic, glass, laminated paper, metal foil, or other materials suitable for holding medicaments.
  • rAAV are provided together with instructions for administering the rAAV to a subject having or at risk of developing the disease or condition (e.g., disease of the CNS, PNS, liver, and the like). Instructions can generally include information about the use of the composition for the treatment or prevention of the disease or condition.
  • a subject having or at risk of developing the disease or condition e.g., disease of the CNS, PNS, liver, and the like.
  • Instructions can generally include information about the use of the composition for the treatment or prevention of the disease or condition.
  • a kit can include allogenic cells.
  • a kit can include cells that can comprise a genomic modification.
  • a kit can comprise“off-the-shelf’ cells.
  • a kit can include cells that can be expanded for clinical use.
  • a kit can contain contents for a research purpose.
  • the instructions include at least one of the following: description of the therapeutic rAAV composition; dosage schedule and administration for treatment or prevention of the disease or condition disclosed herein; precautions; warnings; indications; counter indications; overdosage information; adverse reactions; animal pharmacology; clinical studies; and/or references.
  • the instructions can be printed directly on the container (when present), or as a label applied to the container, or as a separate sheet, pamphlet, card, or folder supplied in or with the container.
  • instructions provide procedures for administering the rAAV to the subject alone.
  • instructions provide procedures for administering the rAAV to the subject at least about 1 hour (hr), 2 hr, 3 hr, 4 hr, 5 hr, 6 hr, 7 hr, 8 hr, 9 hr, 10 hr, 11 hr, 12 hr, 13 hr, 14 hr, 15 hr, 16 hr, 17 hr, 18 hr, 19 hr, 20 hr, 21 hr, 22 hr, 23 hr, 24 hr, 25 hr, 26 hr, 27 hr, 28 hr, 29 hr, 30 hr, or up to 2 days, 3 days, 4 days, 5 days, 6 days, or 7 days after or before administering an additional therapeutic agent disclosed herein.
  • the instructions provide procedures for administering the rAAV to the subject at least about 1 hour (hr), 2 hr, 3 hr, 4 hr, 5 hr, 6 hr, 7
  • instructions provide that the rAAV is formulated for intravenous injection.
  • compositions and methods when used to define compositions and methods, shall mean excluding other elements of any essential significance to the combination for the stated purpose. Thus, a composition consisting essentially of the elements as defined herein would not exclude other materials or steps that do not materially affect the basic and novel characteristic(s) of the claimed disclosure, such as compositions for treating skin disorders like acne, eczema, psoriasis, and rosacea.
  • the terms“homologous,”“homology,” or“percent homology” are used herein to generally mean an amino acid sequence or a nucleic acid sequence having the same, or similar sequence to a reference sequence. Percent homology of sequences can be determined using the most recent version of BLAST, as of the filing date of this application.
  • the terms“increased,” or“increase” are used herein to generally mean an increase by a statically significant amount.
  • the terms“increased,” or“increase,” mean an increase of at least 10% as compared to a reference level, for example an increase of at least about 10%, at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% increase or any increase between 10-100% as compared to a reference level, standard, or control.
  • Other examples of“increase” include an increase of at least 2-fold, at least 5-fold, at least 10-fold, at least 20-fold, at least 50-fold, at least 100-fold, at least 1000-fold or more as compared to a reference level.
  • “decreased” or“decrease” are used herein generally to mean a decrease by a statistically significant amount.
  • “decreased” or“decrease” means a reduction by at least 10% as compared to a reference level, for example a decrease by at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% decrease (e.g., absent level or non-detectable level as compared to a reference level), or any decrease between 10-100% as compared to a reference level.
  • a marker or symptom by these terms is meant a statistically significant decrease in such level.
  • the decrease can be, for example, at least 10%, at least 20%, at least 30%, at least 40% or more, and is preferably down to a level accepted as within the range of normal for an individual without a given disease.
  • the terms“subject” is any organism.
  • the organism is a mammal.
  • mammal include, any member of the mammalian class: humans, non human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice and guinea pigs, and the like.
  • the mammal is a human.
  • the term“animal” as used herein comprises human beings and non-human animals.
  • a“non-human animal” is a mammal, for example a rodent such as rat or a mouse.
  • the subject is a patient, which as used herein, may refer to a subject diagnosed with a particular disease or disorder.
  • the term“gene,” as used herein, refers to a segment of nucleic acid that encodes an individual protein or RNA (also referred to as a“coding sequence” or“coding region”), optionally together with associated regulatory region such as promoter, operator, terminator and the like, which may be located upstream or downstream of the coding sequence.
  • AAV adeno-associated virus
  • AAV AAV type 1
  • AAV2 AAV type 2
  • AAV3 AAV type 3
  • AAV type 4 AAV4
  • AAV type 5 AAV5
  • AAV type 6 AAV6
  • AAV type 7 AAV7
  • AAV type 8 AAV8
  • AAV type 9 AAV9
  • AAV type 10 AAV10
  • AAV type 11 AAV11
  • AAV type 12 AAV 12
  • avian AAV bovine AAV
  • canine AAV equine AAV, primate AAV, non-primate AAV, and ovine AAV.
  • the AAV is described as a“Primate AAV,” which refers to AAV that infect primates. Likewise an AAV may infect bovine animals ( e.g .,“bovine AAV”, and the like). In some instances, the AAV is wildtype, or naturally occurring. In some instances, the AAV is recombinant.
  • AAV capsid refers to a capsid protein or peptide of an adeno-associated virus.
  • the AAV capsid protein is configured to encapsidate genetic information (e.g., a transgene, therapeutic nucleic acid, viral genome).
  • the AAV capsid of the instant disclosure is a modified AAV capsid, relative to a corresponding parental AAV capsid protein.
  • the term“tropism” as used herein refers to a quality or characteristic of the AAV capsid that may include specificity for, and/or an increase or a decrease in efficiency of, expressing the encapsidated genetic information into one in in vivo environment, relative to a second in vivo environment.
  • An in vivo environment in some instances, is a cell-type.
  • An in vivo environment in some instances, is an organ or organ system.
  • the term“AAV vector” as used herein refers to nucleic acid polymer encoding genetic information related to the virus.
  • the AAV vector may be a recombinant AAV vector (rAAV), which refers to an AAV vector generated using recombinatorial genetics methods.
  • rAAV vector comprises at least one heterologous polynucleotide (e.g. a polynucleotide other than a wild-type or naturally occurring AAV genome such as a transgene).
  • the term“AAV particle” as used herein refers to an AAV virus, virion, AAV capsid protein or component thereof. In some cases, the AAV particle is modified relative to a parental AAV particle.
  • the term“gene product” of“gene expression product” refers to an expression product of a polynucleotide sequence such as, for e.g., a polypeptide, peptide, protein or RNA, including interfering RNA (e.g., siRNA, miRNA, shRNA) and messenger RNA (mRNA).
  • operatively linked refers to a location of two or more elements being close together, and in some cases, next to one other (e.g., genetic elements such as a promoter, enhancer, termination signal sequence, polyadenylation sequence, and the like) that enables a functional relationship between the two or more elements.
  • genetic elements such as a promoter, enhancer, termination signal sequence, polyadenylation sequence, and the like
  • a promoter that is operatively linked to a coding region enables the initiation of transcription of the coding sequence.
  • heterologous refers to a genetic element (e.g., coding region) or gene expression product (e.g., RNA, protein) that is derived from a genotypically distinct entity from that of the rest of the entity to which it is being compared.
  • endogenous refers to a genetic element (e.g., coding region) or gene expression product (e.g., RNA, protein) that is naturally occurring in or associated with an organism or a particular cell within the organism.
  • gene expression product e.g., RNA, protein
  • A“detectable moiety” as used herein refers to a moiety that can be covalently or noncovalently attached to a compound or biomolecule that can be detected for instance, using techniques known in the art.
  • the detectable moiety is covalently attached.
  • the detectable moiety may provide for imaging of the attached compound or biomolecule.
  • the detectable moiety may indicate the contacting between two compounds.
  • Exemplary detectable moieties are fluorophores, antibodies, reactive dies, radio-labeled moieties, magnetic contrast agents, and quantum dots.
  • Exemplary fluorophores include fluorescein, rhodamine, GFP, coumarin, FITC, Alexa fluor, Cy3, Cy5, BODIPY, and cyanine dyes.
  • Exemplary radionuclides include Fluorine- 18, Gallium-68, and Copper-64.
  • Exemplary magnetic contrast agents include gadolinium, iron oxide and iron platinum, and manganese.
  • the terms“treat,”“treating,” and“treatment” as used herein refers to alleviating or abrogating a disorder, disease, or condition; or one or more of the symptoms associated with the disorder, disease, or condition; or alleviating or eradicating a cause of the disorder, disease, or condition itself. Desirable effects of treatment can include, but are not limited to, preventing occurrence or recurrence of disease, alleviation of symptoms, diminishing any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state and remission or improved prognosis.
  • terapéuticaally effective amount refers to the amount of a compound or therapy that, when administered, is sufficient to prevent development of, or alleviate to some extent, one or more of the symptoms of a disorder, disease, or condition of the disease; or the amount of a compound that is sufficient to elicit biological or medical response of a cell, tissue, system, animal, or human that is being sought by a researcher, veterinarian, medical doctor, or clinician.
  • the term“pharmaceutically acceptable carrier,”“pharmaceutically acceptable excipient,”“physiologically acceptable carrier,” or“physiologically acceptable excipient” refers to a pharmaceutically acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, excipient, solvent, or encapsulating material.
  • a component can be“pharmaceutically acceptable” in the sense of being compatible with the other ingredients of a pharmaceutical formulation. It can also be suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio. See, Remington: The Science and Practice of Pharmacy, 21st Edition; Lippincott Williams & Wilkins:
  • composition refers to a mixture of a compound disclosed herein with other chemical components, such as diluents or carriers.
  • the pharmaceutical composition can facilitate administration of the compound to an organism. Multiple techniques of administering a compound exist in the art including, but not limited to, oral, injection, aerosol, parenteral, and topical administration.
  • Non-limiting examples of“sample” include any material from which nucleic acids and/or proteins can be obtained. As non-limiting examples, this includes whole blood, peripheral blood, plasma, serum, saliva, mucus, urine, semen, lymph, fecal extract, cheek swab, cells or other bodily fluid or tissue, including but not limited to tissue obtained through surgical biopsy or surgical resection.
  • the sample comprises tissue from the large and/or small intestine.
  • the large intestine sample comprises the cecum, colon (the ascending colon, the transverse colon, the descending colon, and the sigmoid colon), rectum and/or the anal canal.
  • the small intestine sample comprises the duodenum, jejunum, and/or the ileum.
  • a sample can be obtained through primary patient derived cell lines, or archived patient samples in the form of preserved samples, or fresh frozen samples.
  • the term“ vivo” is used to describe an event that takes place in a subject’s body.
  • ex vivo is used to describe an event that takes place outside of a subject’s body.
  • An ex vivo assay is not performed on a subject. Rather, it is performed upon a sample separate from a subject.
  • An example of an ex vivo assay performed on a sample is an“m vitro” assay.
  • vitro is used to describe an event that takes places contained in a container for holding laboratory reagent such that it is separated from the biological source from which the material is obtained.
  • In vitro assays can encompass cell-based assays in which living or dead cells are employed.
  • In vitro assays can also encompass a cell-free assay in which no intact cells are employed.
  • Example 1 Method of Producing an rAAV
  • a recombinant AAV (rAAV) is produced.
  • Three plasmid vectors are triple- transfected into an immortalized HEK293 using a standard transfection protocol (e.g ., with PEI).
  • the first vector contains a transgene cassette flanked by inverted terminal repeat (ITR) sequences from a parental AAV virus.
  • the transgene cassette has a promoter sequence and that drives transcription of a heterologous nucleic acid in the nucleus of the target cell.
  • the second vector contains nucleic acids encoding the AAV Rep gene, as well as a modified Cap gene e.g., AAV2/9 REP-AAP-ACap).
  • Modified Cap gene comprises any one of the DNA sequences provided in FIGS.
  • the third vector contains nucleic acids encoding helper virus proteins needed for viral assembly, and packaging of the heterologous nucleic acid into the modified capsid structure.
  • Viral particles are harvested from the media after 72 h post transfection and from the cells and media at 120 h post transfection.
  • Virus present in the media is concentrated by precipitation with 8% poly(ethylene glycol) and 500 mM sodium chloride and the precipitated vims is added to the lysates prepared from the collected cells.
  • the viruses are purified over iodixanol (Optiprep, Sigma) step gradients (15%, 25%, 40% and 60%).
  • Viruses are concentrated and formulated in PBS. Vims titers are determined by measuring the number of DNasel-resistant vector genome copies (VGs) using qPCR and the linearized genome plasmid as a control.
  • VGs DNasel-resistant vector genome copies
  • Example 2 Methods of Identifying Variant AAV Capsid Proteins
  • the rAAV-ACap-in-cis-Lox2 plasmid (FIG. 36) is a modification of the rAAV-ACap-in-cis-Lox plasmid.
  • the pCRII- 9Cap-XE plasmid was used as a template.
  • the AAV2/9 REP-AAP-ACap plasmid (FIG. 36) was modified from the AAV2/9 REP-AAP plasmid.
  • the rAAV-ACap-in-cis-Lox2 plasmid consists of three major elements that are flanked by AAV2 ITRs.
  • the mCherry expression cassette of the previous version of the plasmid was replaced by mNeonGreen cassette
  • a portion of AAV2 rep gene that has the splicing sequences and AAV5 p41 promoter (1680- 1974 residues of GenBank AF085716.1) followed by AAV9 cap gene.
  • the prior version of this plasmid has a short 12 bp sequence between restriction sites Xbal and Agel at AA 450 and 592 of the AAV9 Cap gene. This was replaced by a 723 bp sequence of mRuby2 gene in-frame (acts as filler DNA) in the newer version of the plasmid (iii) SV40 polyadenylation sequence that is flanked by lox71 and lox66 sites. The minor changes were introduced to the prior version of the plasmid to facilitate ease of cloning and to visualize mammalian cell transfection. The Lox sites in these rAAV plasmids show modest levels of Cre- independent flipping.
  • the pCRIT9Cap-XE plasmid contains the AAV9 capsid gene sequence from AAs 450-592 and is flanked by Xbal and Agel restriction sites.
  • the AAV2/9 REP-AAP-ACap plasmid has the five previously existing stop codons of AAV2/9 REP-AAP in addition to the deletion of AAs 450-592 of the AAV9 capsid sequence. These modifications did not affect vector production.
  • the deletion of the overlapping fragment between the REP-AAP and rAAV-ACap-in-cis-Lox2 plasmids minimizes recombination between plasmids that could potentially generate AAV9 wild-type capsids during co-transfection in vector production.
  • AAV capsids The AAV capsid variants with 7-mer insertions or 11-mer substitutions were made between positions 587-597 of AAV-PHP.B capsid using the pUCmini-iCAP-PHP.B backbone (Addgene ID: 103002).
  • ssAAV genomes To characterize the AAV capsid variants, the single stranded (ss) rAAV genomes were used. Genomes such as pAAV:CAG-mNeonGreen27 (equivalent plasmid, pAAV: CAG-eYFP35; Addgene ID: 104055), pAAV:CAG-NLS-EGFP26 (equivalent version with one NLS is on Addgene ID 104061), pAAV:CAG-DIO-EYFP35 (Addgene ID: 104052), pAAV: GfABC lD-2xNLS-mTurquoise235 ( Addgene ID: 104053), and pAAV-Ple261-iCre30 (Addgene ID 49113) were used.
  • pAAV:CAG-mNeonGreen2 genome consists of a ubiquitous CMV-P-Actin-intron-P- Globin (CAG) hybrid promoter driving the expression of a fluorescent protein, mNeonGreen (equivalent plasmid, pAAV: CAG-eYFP3; Addgene ID: 104055).
  • pAAV:CAG-NLS-EGFPl consists of NLS sequences at the N- and C-termini of EGFP and is driven by the CAG promoter. An equivalent version with one NLS is on Addgene (ID 104061).
  • pAAV:CAG-DIO-EYFP3 (Addgene ID: 104052) consists of a EYFP gene built in the reverse direction of the CAG promoter, and it is flanked by a pair of Cre-Lox sites (Lox P and Lox 2272) on either ends.
  • pAAV GfABC 1D- 2xNLS-mTurquoise23, referred to elsewhere as pAAV:GFAP-2xNLS-mTurquoise2 ( Addgene ID: 104053), consists of NLS sequences at the N- and C-termini of mTurquoise2 and is driven by the astrocyte-specific promoter GfABClD4.
  • pAAV:Ple261-iCre5 contains an endothelial-cell-specific promoter driving the expression of iCre.
  • pAAV:CAG-XFP (mNeongreen) was packaged for characterizing AAV variants. However, when performing quantification of cell-types: neurons, astrocytes and
  • CAG-NLS-EGFP was used to restrict the expression to nucleus for easier quantification using microscope images.
  • GFAP-NLS-mTurq2 is used to quantify astrocytes.
  • CAG-DIO-EYFP is used for Cre driver lines, due to the presence of lox sites in this plasmid.
  • the self-complementary genome from Dr. Guangping Gao, scAAV:CB6-EGFP genome has a hybrid ubiquitous CB6 promoter (975 bp) comprising a CMV enhancer
  • cytomegalovirus immediate early enhancer a chicken-P-actin promoter and hybrid intron, that drives the expression of EGFP.
  • the genome has a rabbit globin poly A (127 bp) following the EGFP gene.
  • the scAAV:CAG-EGFP (Addgene ID:83279), vector uses a ubiquitous CMV-b- Actin-intron-P-Globin (CAG) hybrid promoter to drive the expression of EGFP.
  • N can be A, C, G, or T bases and K can be G, or T.
  • N can be A, C, G, or T bases and K can be G, or T.
  • PCR amplification was limited to 15 - 20 cycles and the reactions were scaled up to get the required yield.
  • the resulting PCR products were run on a 1% agarose gel and extracted with a Zymoclean Gel DNA Recovery kit (Zymo Research; D4007). It is critical to avoid AAV contamination during this step by taking precautionary measures like using a clean gel-running box and freshly prepared lx TAE buffer.
  • rAAV-ACap-in-cis-Lox2 plasmid (6960 bp) was linearized with the restriction enzymes Agel and Xbal by following the NEB recommended protocol for double digestion.
  • the digested plasmid was run on a 0.8% - 1% agarose gel to extract the linearized backbone (6237 bp) with a Zymoclean Gel DNA Recovery kit.
  • the amplified library fragment was assembled into the linearized vector with the NEBuilder HiFi DNA Assembly Master Mix (NEB; E2621S) and a 1:2 molar ratio of vector to insert, to assemble at 50°C for 60 min.
  • NEB NEBuilder HiFi DNA Assembly Master Mix
  • This area is then used to determine the fraction of an individual library that needs to be pooled into PCR pool library using the formula: [Area under the curve/ total number of libraries pooled] .
  • the pooled sample was used as a template for further amplification with 12 cycles of 98°C for 10 s, 60°C for 20 s, and 72°C for 30 s by Q5 polymerase, using the primers 588-R21ib- F: 5’-CACTCATCGACCAATACTTGTACTATCTCTCT-3’ and 588-R21ib-R: 5’- GTATTCCTTGGTTTTGAACCCAACCG-3’ . Similar to R1 library generation, the PCR product was assembled into the rAAV-ACap-in-cis-Lox2 plasmid and the virus was produced.
  • the R1 libraries used to build R2 were the Cre-Lox flipped rAAV DNA from half of the mouse brains (-0.3 g) and portion of spinal cords (0.1-0.2g) from all Cre lines. The amount of tissue processed here was sufficient for complete capsid library recovery.
  • the differentially pooled and amplified libraries (by PCR pool or synthetic pool) were assembled using gibson assembly with a follow-up PS or Exonuclease V treatment (as described in R1 library generation). Successful library generation was validated by transformation, Sanger sequencing, and an ITR Smal digest.
  • spike-in library has 11-mer mutated variants
  • the same primer design where“XXXXXMNNMNNMNNMNNMNNMNNMNNMNNXXXX” was replaced with a specific nucleotide sequence of a 11-mer variant.
  • a duplicate of each sequence in this library was designed with different codons optimized for mammals.
  • the primers were designed using a custom built python based script (code will be made available in Github).
  • the custom-designed oligopool was synthesized in an equimolar ratio by Twist Biosciences.
  • the oligopool was used to amplify the pCRITXE Cap9 template over 13 cycles of 98°C for 10 s, 60°C for 20 s, and 72°C for 30 s.
  • the product of the first PCR was used as a template for the second PCR using the primers XF and 588-R21ib-R (described above) and amplified for 13 cycles.
  • the PCR product was assembled into an rAAV backbone and processed and purified for virus production. About 10 ng per 150 mm dish of HEK293 cells produced about 6xlO u vg of virus library.
  • the plasmid pUC18 acts as a filler DNA to compensate for the low amount of library DNA in order to maintain the N:P ratio required for optimal transfection using polyethylenimine (PEI, Polysciences; 24765-1) transfection).
  • PEI polyethylenimine
  • the cells and culture media were harvested at 60 h post transfection to collect the viral particles. rAAV harvest and purification were performed as per the protocol. The small amount of library DNA per plate and early cell harvest time are critical for reducing the possibility of mosaic capsid assemblies during vector production (similar considerations seen in prior reports).
  • rAAV DNA extraction from purified rAAV viral library -10% of the purified viral library was used to extract the viral genome by proteinase K treatment.
  • DNase I enzyme (5 pi of 10 U/pl) (Sigma-Aldrich; 4716728001) in 100 m ⁇ of DNase I buffer and incubated for 1 h at 37°C. The enzyme was inactivated by adding 5 m ⁇ of 0.5 M EDTA at 70°C for 10 min.
  • the capsid protein shell was digested by adding 120 m ⁇ of proteinase solution containing 5 m ⁇ of 20 pg/m 1 of proteinase K and incubated at 50°C overnight. To inactivate the proteinase K, the mixture was boiled at 95°C. The extracted rAAV library DNA was then concentrated and purified using phenol chloroform and ethanol. An equal volume of PhenohChloroformTsoamyl Alcohol 25:24:1, pH8.0 (-250 m ⁇ ; ThermoFisher Scientific; 15593031) was added and vortexed for 30 s. The mixture is incubated for 5 min at room temperature (RT) before centrifugation at 15,000 rpm for 10 min at 4°C. The upper aqueous phase was separated and mixed with an equal volume of chloroform and vortexed for 30 s.
  • RT room temperature
  • mice lines used in this study were purchased from the Jackson Laboratory (JAX).
  • JX Jackson Laboratory
  • 6- to 8-week-old adult male and female mice were intravenously injected with the viral libraries. Both genders were used for capsid selection to recover capsid variants with minimal gender bias.
  • the IV injection of rAAVs was into the retro-orbital sinus of adult mice.
  • mice were euthanized and all organs including brain were collected, snap frozen on dry ice, and stored at -80°C.
  • the total rAAV genome recovery from 0.1 g of mouse liver was quantified by quantitative PCR using the primers mNeonGreen-F: 5’- CG AC AC AT G AGTT AC AC AT CTTT GGCTC- 3’ and mNeonGreen-R: 5’- GGAGGTC ACCCTTGGTGGACTTC-3’ , which binds to the mNeonGreen gene of the ssAAV- ACap-in-cis-Lox2 genome.
  • the amount of mitochondrial DNA was quantified using primers Mito-F: 5’- CCCAGCTACTACCATCATTCAAGT-3’ and Mito-R: 5’-
  • the extracted viral genome was digested with a restriction enzyme, such as Smal (found within the ITRs), to improve rAAV genome recovery by PCR.
  • a restriction enzyme such as Smal (found within the ITRs)
  • rAAV genome extraction with the Trizol method Half of a frozen brain hemisphere (0.3 g approx.) was homogenized with a 2 ml glass homogenizer (Sigma Aldrich; D8938) or a motorized plastic pestle (Fisher Scientific;12-141-361, 12-141-363) (for smaller tissues) and processed as described in prior work. The extracted DNA was then treated with 3 - 6 pi of 10 pg/m ⁇ RNase Cocktail Enzyme Mix (ThermoFisher Scientific; AM2286) to remove RNA and digested with Smal restriction enzyme. The treated mixture was then purified with a Zymo DNA Clean and Concentrator kit (D4033). From deep sequencing data analysis, it was observed that the amount of tissue processed for rAAV genome recovery is sufficient.
  • rAAV genome recovery by Cre-dependent PCR rAAV genomes with Lox sites flipped by Cre recombination were selectively recovered and amplified using PCR with primers that yield a PCR product only if the Lox sites are flipped (See FIG. 37).
  • CAAGTAAAACCTCTACAAATGTGGTAAAATCG-3’ were used and amplified the Cre- recombined genomes over 25 cycles of 98°C for 10 s, 58°C for 30 s, and 72°C for 1 min, using Q5 DNA polymerase
  • ACTCATCGACCAATACTTGTACTATCTCTCTAGAAC-3’ 588-R21ib-R (5’- GTATTCCTTGGTTTTGAACCCAACCG-3’) were used to amplify the genomes over 25 cycles of 98°C for 10 s, 60°C for 30 s, and 72°C for 30 min, using Q5 DNA polymerase.
  • GTGACTGGAGTTCAGACGTGTGCTCTTCCGATCTTCCTTGGTTTTTTGAACCCAACCG-3’ that are positioned around 50 bases from the randomized 7-mer insertion on the capsid, and that contain the Readl and Read2 flow cell sequences on the 5’ end.
  • CACGACGCTCTTCCGATCTAANNNNNNAGTCCTATGGAC AAGTGGCC ACA-3’ used to minimally amplify DNA and virus libraries for NGS has 6 nucleotides long UID (unique identifier)“NNNNNN” that sits after 19 nucleotides of Read-1 sequence used in NGS“5’- CACGACGCTCTTCCGATCT” and linker“AA”. The sequence after UID
  • AGTCCTATGGACAAGTGGCCACA is the region that anneals to the AAV9 capsid.
  • UID is an optional feature for NGS data analysis to identify potential PCR amplification errors.
  • PCR products post indices addition were run on a freshly prepared 2% low- melting-point agarose gel (ThermoFisher Scientific; 16520050) for better separation and recovery of the approx. 120 bp DNA band on the gel.
  • the nucleotide diversity at the randomized 7-mer position was verified by Sanger sequencing. If needed, an optional PCR was carried out to send sufficient sample for Sanger sequencing using 15 - 20 cycles of 98°C for 10 s, 60°C for 30 s, and 72°C for 10 s with the primers NGS-QC-F:
  • CAAGCAGAAGACGGCATACGA-3 The libraries were sent for deep sequencing using the Illumina HiSeq 2500 System (Millard and Muriel Jacobs Genetics and Genomics Laboratory, Caltech; Integrative Genomics Core, City of Hope).
  • the DNA was amplified by Q5 Hot Start High-Fidelity 2X Master Mix or NEBNext Ultra II Q5 Master Mix (NEB; M0544) for 10 cycles of 98°C for 10 s, 59°C for 30 s, and 72°C for 10 s.
  • the mixture was purified with a PCR purification kit.
  • the eluted DNA was then used as a template in a second PCR to add the unique indices (single or dual) using the recommended primers (NEB; E7335S, E7500S, E7600S) in a 10-cycle reaction with the same temperature cycle as described above (for DNA and vims library preparation).
  • the extracted DNA was validated by Sanger sequencing and sent for deep sequencing as described in the previous section.
  • AAV capsid variants Cloning AAV capsid variants.
  • the AAV capsid variants were cloned into a pUCmini- iCAP-PHP.B backbone (Addgene ID: 103002) using overlapping forward and reverse primers with 11-mer substitution (in case of 7-mer-i variants, the flanking amino acids from AAV9 capsid AA587-588“AQ” and AA589-590“AQ” were subjected to codon modification) that spans from the Mscl site (at position 581 AA) to the Agel site (at position 600 AA) on the pUCmini plasmid.
  • the primers were designed for all capsid variants using a custom python script (Code will be made available on github), and since they cover the entire fragment insertion, these primers are self-annealed and amplified using PCR to create a dsDNA fragment without the use of a template DNA. They are amplified by Q5 Hot Start High-Fidelity 2X Master Mix for 20 cycles of 98°C for 10 s, 60°C for 30 s, and 72°C for 10 s. This fragment was then assembled into the scl/Agcl digested pUCmini-iCAP-PHP.B backbone by the Gibson assembly method. There is a second Mscl site on the backbone; however, this was blocked by methylation. The assembled plasmids were then transformed into NEB Stable Competent E. coli (New England Biolabs, Inc; C3040H), and colonies were selected on carbenicillin/ampicillin-LB agar plates.
  • AAV vector production Using an optimized protocol, AAV vectors were produced from 5 -10 150 mm plates, which yielded sufficient amounts for administration to adult mice.
  • AAV vector administration AAV vectors were administered intravenously to adult male mice (6 - 8 weeks of age) via retro-orbital injection at doses of 1 - lOxlO 11 vg.
  • the AAV doses are determined by the experimental needs.
  • CAG-NLS-GFP related experiments for quantification were done at medium dose of lxlO 11 vg given this was the dose previously determined for AAV-PHP.eB characterization. Otherwise, the non-NLS genome related experiments were done at 3xl0 n vg, with the exception of Cre-driver lines (GFAP-Cre or Tek- Cre), or a lower strength promoter containing genome (GFAP-NLS-mTurq) where the dose was 1x10 vg.
  • the high dose was chosen to understand the full potential of the new vectors in these systems.
  • mice were anesthetized with Euthasol (pentobarbital sodium and phenytoin sodium solution, Virbac AH) and transcardially perfused with 30 - 50 mL of 0.1 M phosphate buffered saline (PBS) (pH 7.4), followed by 30 - 50 ml of 4% paraformaldehyde (PFA) in 0.1 M PBS. After this procedure, all organs were harvested and post-fixed in 4% PFA at 4°C overnight. The tissues were then washed and stored at 4°C in 0.1 M PBS and 0.05% sodium azide. All solutions used for this procedure were freshly prepared. For the brain and liver, lOO-pm thick sections were cut on a Leica VT1200 vibratome.
  • Euthasol pentobarbital sodium and phenytoin sodium solution, Virbac AH
  • PFA paraformaldehyde
  • mice were anesthetized and transcardially perfused with 20 mL of ice-cold PBS, followed by 10 mL of ice-cold PBS containing Texas Red-labeled
  • Lycopersicon Esculentum (Tomato) Lectin (1:100, Vector laboratories, TL-1176), and then placed in 30 mL of ice-cold 4% PFA for fixation.
  • the tissues were washed 1 - 3 times with wash buffer 1 (0.1% Triton X-100 in 0.1 M PBS buffer, pH 7.4) over a period of 5 - 6 h in total.
  • the tissues were then incubated in blocking buffer with the secondary antibodies at appropriate dilutions for 12 - 24 h at room temperature and then washed in three times in 0.1 M PBS, pH 7.4 over a total duration of 5 - 6 h.
  • the secondary antibody used in this study was Alexa Fluor 647 AffiniPure donkey anti-rabbit IgG (H+L) (Jackson ImmunoResearch Lab, 711-605-152).
  • DAPI Diamidine-2'-phenylindole dihydrochloride
  • Hybridization chain reaction (HCR) based RNA labeling in tissues.
  • Fluorescence in situ hybridization chain reaction (FITC-HCR) was used to label excitatory neurons with VGLUT1 and inhibitory neurons with GAD1 to characterize the AAV capsid variant AAV- PHP.N in brain tissue using an adapted third-generation HCR protocol.
  • HCR method was sought to label excitatory and inhibitory neurons.
  • Fluorescence in situ hybridization chain reaction (FITC-HCR) was used to label excitatory neurons with VGLUT1 and inhibitory neurons with GAD1.
  • Tissue slices were permeabilized with 0.1% Triton X-100 in 0.1 M RNase-free PBS for 1 h at RT and pre-hybridized in hybridization solution (10% dextran sulfate and 10% ethylene carbonate in 2xSSC buffer (saline-sodium citrate)) for >30 min at 37°C.
  • hybridization solution 10% dextran sulfate and 10% ethylene carbonate in 2xSSC buffer (saline-sodium citrate)
  • the designed probes were diluted in hybridization solution to get a final concentration of 2 nM.
  • the tissue sections were then subjected to hybridization with the probes overnight at 37°C.
  • Imaging and image processing All images in this study were acquired either with a Zeiss LSM 880 confocal microscope using the objectives Fluar 5x 0.25 M27, Plan-Apochromat lOx 0.45 M27 (working distance 2.0 mm), and Plan-Apochromat 25x 0.8 Imm Corr DIC M27 multi-immersion; or with a Keyence BZ-X700 microscope.
  • the acquired images were processed in Zen Black 2.3 SP1 (Zeiss), BZ-X Analyzer (Keyence), Illustrator CC 2018 (Adobe),
  • the brain hemisphere was stained with the primary antibody, Anti-GFP (Aves Labs, GFP-1020), and the secondary antibody, goat anti-Chicken IgY, Alexa Fluor 633 (ThermoFisher Scientific, A-21103), and cleared via the iDISCO protocol .
  • Anti-GFP Administered Immunosorbent
  • goat anti-Chicken IgY Alexa Fluor 633
  • Alexa Fluor 633 ThermoFisher Scientific, A-21103
  • Tissue processing and imaging for quantification of rAAV transduction in vivo 6- to 8-week-old male mice were intravenously injected with the vims, which was allowed to express for 3 weeks (unless specified otherwise). The mice were randomly assigned to groups and the experimenter was not blinded. The mice were perfused and the organs were fixed in PFA. The brains and livers were cut into lOO-pm thick sections and immunostained with different cell-type- specific antibodies, as described above. The images were acquired either with a 25x objective on a Zeiss LSM 880 confocal microscope or with a Keyence BZ-X700 microscope; images that are compared directly across groups were acquired and processed with the same microscope and settings.
  • HBMEC Human Brain Microvascular Endothelial Cells
  • the cells were cultured from a frozen stock vial in fibronectin-coated T-75 flask (7000-9000 cells/cm seeding density) using the Endothelial Cell Medium (Cat. 1001).
  • the cells were subcultured in fibronectin-coated 48-well plates (0.95 cm growth area) at the recommended seeding density and incubated at 37°C for ⁇ 24-48 h till the cells were completely adherent with ⁇ 70-80% confluence.
  • the viral vectors packaging pAAV-CAG-mNeongreen were added to the cell culture at a dose of either 1x108 or 1x1010 vg per well (3 wells per dose per vector).
  • the media was changed 24 hours later and the culture was assessed for fluorescence expression at 3 days’ post infection. Per vendor recommendation, the culture media was changed every other day to maintain the cell culture.
  • NGS data alignment and processing The raw fastq files from NGS runs were processed with custom built scripts (codes will be made available on Github) that align the data to AAV9 template DNA fragment containing the diversified region 7xNNK (for Rl) or 1 lxNNN (for R2 since it was synthesized as 1 lxNNN).
  • the pipeline to process these datasets involved filtering the dataset to remove the low-quality reads by using the deep sequencing quality score for each sequence.
  • the variant sequences were then recovered from the sequencing reads by searching for the flanking template sequences, and extracting the nucleotides of the diversified region (perfect string match algorithm).
  • the quality of the aligned data was further investigated to remove any erroneous sequences (such as ones with stop codons).
  • the raw data was plotted (as shown in Supplementary Fig. le) to study the quality of recovery across every library.
  • a thresholding method to remove plausible erroneous mutants that may have resulted from PCR or NGS based errors. The assumption is that if there is a PCR mutation or NGS error on the recovered parent sequence, the parent must have existed at least one round earlier than the erroneous sequence, and thus a difference in RCs should exist.
  • This step partially removed the long tail of low count reads.
  • the thresholding that was described for Rl tissue libraries was applied.
  • Standard score (read count_i - mean)/standard deviation. Where read count_i is raw copy number of a variant i, Mean is the mean of read counts of all variants across a specific library, Standard deviation is the standard deviation of read counts of all variants across a specific library.
  • R1 virus library is the input library to the R1 tissue libraries.
  • the estimated RC is defined as a number that is lower than the lowest RC in the library with the assumption that these variants were found at a relatively lower abundance than the variants recovered from the deep sequencing.
  • RC of 1.0 was the lowest, we assigned all missing variants an estimated RC of 0.9.
  • Fig. Id we use this method to calculate the enrichment score of the R1 tissue libraries which is normalized to R1 virus library (Fig. Id). This was done to represent libraries across two selection rounds consistently.
  • the individual enrichment score among R1 variants didn’t add a significant value to the variants selected for R2 selection as described in the criteria to separate signal vs noise in R1 using the RCs.
  • Heatmap generation The relative AA distributions of the diversified regions are plotted as heatmap s.
  • the plots were generated using the Python Plotly plotting library.
  • the heatmap values were generated from custom scripts written in Python, using functions in the custom“pepars” Python package.
  • Each heatmap uses both an expected (input) distribution of amino acid sequences and an output distribution.
  • the output distribution must be a list of sequences and their count, and the input distribution can be either a list of sequences and their count, or an expected amino acid frequency from a template, such as NNK.
  • the total count of amino acids in each position is tallied in accordance to each sequence's count and then divided by the total sum of counts, giving a frequency of each amino acid at each position.
  • the log2 fold change is calculated between the output and the input. For amino acids with a count of 0 in either the input or output, no calculation is performed. In order to distinguish between statistically significant amino acid biases, a statistical test was performed using the statsmodels Python library. For the case where there are two amino acid counts, a two- sided, two-proportion z-test was performed; for comparing the output amino acid count to an expected input frequency from a template, a one-proportion z-test was performed. All p-values were then corrected for multiple comparisons using Bonferroni correction. Only bias differences below a significance threshold of le-4 are then outlined on the heatmap; all other (insignificant) squares are left empty.
  • the number on the bottom represents the position of the diversified motif starting from 1.
  • the size of the amino acid in the stack reflects the proportion of unique clones in which the AA appears at that specific position in the motif.
  • the color code is based on the AA properties.
  • the positively charged residues K, R, and H are in blue.
  • the negatively charged residues D and E are in red.
  • the amide containing polar residues Q, and N are in magenta.
  • the polar residues T, and S, are in green.
  • the hydrophobic residues A, L, V, I, P, F, M, and W are in black.
  • Example 3 Multiplexed-CREATE allows detailed characterization of the capsid libraries during Round- 1 selection.
  • mice per Cre transgenic line see Methods.
  • IV intravenous
  • the brain and liver tissues were harvested, with the latter serving as a control organ since AAV9 transduces it with high efficiency.
  • the rAAV genomes were extracted from tissues and the capsids that transduced Cre-expressing cells were selectively amplified (FIGS. 40-44).
  • FIGS. 40-44 Upon deep sequencing, ⁇ 8xl0 4 unique nucleotide variants recovered from brain tissues and ⁇ 50 variants in spinal cords (-48% of which were identified in vims library) were observed across the transgenic lines, and each variant was represented with an enrichment score reflecting the
  • Example 4 A novel Round-2 library design improves the selection outcome
  • Table 7 Comparison between the two methods for R2 selection. The table summarizes the pros and cons of selection design parameters by the synthetic pool and PCR pool R2 selection methods.
  • the synthetic pool library design comprised: (1) equimolar amounts of -8950 capsid variants present at high read counts in at least one of the R1 selections from brain and spinal cord (FIG. 40); (2) alternative codon replicates of those -8950 variants (optimized for mammalian codons) to reduce false positives; and (3) a“ spike-in” library of controls (FIGS. 74 and 75), resulting in a total library size of 18,000 nucleotide variants.
  • both round-2 (R2) virus libraries produced a high titer ( ⁇ 6xlO u vg per 10 ng of R2 DNA library per 150 mm dish; FIG. 45), and -99% of variants from the R2 DNA were found after viral production (FIG. 6).
  • the PCR pool library carries forward the R1 selection biases (FIGS. 7, 46, and 47) where the abundance reflects prior enrichment across tissues in R1 as well as bias from viral production and sample mixing. Comparatively, the synthetic pool DNA library is more evenly distributed, minimizing bias amplification across selection rounds.
  • rAAV genomes from brain samples were extracted, selectively amplified, and deep sequenced (as in Rl).
  • the synthetic pool library produced a greater number of positively enriched capsid variants than the PCR pool brain library (e.g. -1700 versus -700 variants/tissue library at amino acid (AA) level in GFAP-Cre) (FIGS. 8 and 48).
  • AA amino acid
  • PCR pool design coupled with enrichment normalization to virus library may not drastically differ from synthetic pool design over one additional round of selection for a subset of in vivo selections (such as Tek-Cre or SNAP-Cre). Without additional validation, however, it is difficult to predict whether a given in vivo system will perform akin to Tek-Cre. This becomes critical in a multiplexed selection study where target- specific variants may not garner the highest enrichments in one particular in vivo selection.
  • Table 8 Ranking of AAV-PHP capsids across methods. Ranks of selected variants among all capsids recovered from R2 Tek-Cre selection by synthetic pool enrichment score (representing M-CREATE), PCR pool enrichment score (representing closer to M-CREATE), or PCR pool read counts (representing CREATE), the highest ranks of which starts from 1, and “Not recovered” represent absence of the variant from R2 sequencing data.
  • Example 6 Capsid recovery from Round-2 selection yields a pool of AAV9 variants with enhanced BBB entry and CNS transduction [0377] Given the dominance of the PHP.B-family in this particular selection, the most enriched member was tested: TALKPFL (FIGS. 12 and 13) henceforth referred to as AAV- PHP.V1. Somewhat surprisingly given its sequence similarity to AAV.PHP.B, the tropism of AAV-PHP.V1 is biased toward transducing brain vascular cells (FIGS. 14, 54).
  • AAV-PHP.V1 When delivered intravenously, AAV-PHP.V1 carrying a fluorescent reporter under the control of the ubiquitous CAG promoter transduces -60% of GLUT1 + cortical brain vasculature compared to -20% with AAV-PHP.eB and almost no transduction with AAV9 (FIGS. 14 and 16). In addition to the vasculature, AAV-PHP.V1 also transduced -60% of cortical S100 + astrocytes (FIG. 17).
  • AAV-PHP.V1 is not as efficient for astrocyte transduction as the previously reported AAV-PHP.eB (when packaged with an astrocyte specific GfABCID promoter, FIG. 55).
  • AAV-PHP.V1 vectors can be used in three different systems: (1) in endothelial cell- type specific Tek-Cre mice with a Cre-dependent expression vector (FIG. 15 (left) and FIG. 18), (2) in fluorescent reporter mice where Cre is delivered with an endothelial cell-type specific MiniPromoter (Ple261) (FIG. 15 (right) and FIGS. 19 and 56 through the left column of FIG.
  • AAV-PHP.V2 - TTLKPFL differed by only one AA from AAV-PHP.V1
  • AAV-PHP.V2 was found at high abundance in R1 selection across all brain libraries and was highly enriched in R2 (FIGS. 4, 11 (right panel), 12, 13, and 40). Given its sequence similarity, similar tropism was expected to that of AAV-PHP.V1.
  • AAV-PHP.B6 - TLQLPFK and AAV-PHP.B7 - TLQQPFK (FIGS. 48 (middle panel), and 53) that were previously identified in the 3-mer-s PHP.B library but never validated in vivo.
  • these variants also display PHP.B-like tropism (FIGS. 20-21 and 62).
  • a series of variants selected to verify M-CREATE’s predictive power outside this family were then investigated: (1) A highly enriched variant with a completely unrelated sequence, AAV-PHP.C1 - RYQGDSV (FIGS. 12, 13, 20, and 21), transduced astrocytes at a similar efficiency and neurons at lower efficiency compared to other tested variants from B- family (FIG. 21). (2) Two variants found in high abundance in the R2 synthetic pool vims library and negatively enriched in brain (with both codon replicates in agreement), AAV-PHP.X1 - ARQMDLS and AAV-PHP.X2 - TNKVGNI (FIG. 46, right), poorly transduced the CNS (FIG. 63).
  • Example 7 Re-investigation of capsid selection that yielded AAV.PHP.eB reveals variant that specifically transduces neurons
  • the re-investigated 3- mer-s PHP.B library diversified positions 587-597 of the AAV-PHP.B capsid (equivalent of 587- 590 AA on AAV9) in portions of three consecutive AAs, (-40,000 total variants) (FIG. 24). Selections were performed in three Cre-transgenic lines: Vglut2-IRES-Cre for glutamatergic neurons, Vgat-IRES-Cre for GABAergic neurons, and GFAP-Cre for astrocytes.
  • Variants that were positively enriched in brain and negatively enriched in liver show a significant bias towards certain AAs: G, D, E at position 1; G, S at position 2 (which includes the AAV-PHP.eB motif, DG); and S, N, P at position 9, 10, 11 (FIGS. 26 and 68).
  • Variants that were positively enriched in the brain were clustered according to their sequence similarities and ranked by their negative enrichment in liver (represented by node size in clusters).
  • a distinct family referred to as N emerged with a common motif“SNP” at positions 9-11 on PHP.B backbone (FIGS. 27 and 69).
  • the core variant of the N-family cluster was found in high abundance in R1 and R2 selections, had higher enrichment score in Vglut2 and Vgat brain tissues compared to GFAP, and had negative enrichment in liver tissue (FIGS. 25 and 66-69).
  • this variant specifically transduced NeuN + neurons even when packaged with a ubiquitous CAG promoter, although the transduction efficiency varied across brain regions (from -10-70% in NeuN + neurons, including both VGFUT1 + excitatory and GAD1 + inhibitory neurons, FIGS. 28, 29, 70, and 71).
  • AAV-PHP.eB The enhanced CNS tropism of AAV-PHP.eB is absent in a subset of mouse strains. It is highly efficient in C57BL/6J, FVB/NCrl, DBA/2, and SJL/J, with intermediate enhancement in 129Sl/SvimJ, and no enhancement in BALB/cJ and several additional strains. This pattern holds for the two newly identified variants from the PHP.B family, AAV-PHP.V1 and AAV- PHP.N (FIG. 30, Table 9), which did not transduce the CNS in BALB/cJ, yet transduced the FVB/NJ strain (FIG. 31).
  • AAV-PHP.V1 transduced Human Brain Microvascular Endothelial Cell (HBMEC) culture, resulting in increased mean fluorescent intensity compared to AAV9 and AAV-PHP.eB (FIG. 72) however, suggesting the potential for mechanistic complexity.
  • Table 9 AAV-PHP vectors identified by CREATE and M-CREATE. The table provides a summary of the variants that have been identified so far using CREATE and M- CREATE, along with their tropism and the evolutionary steps from the parent capsid that was involved in their discovery.
  • M-CREATE revealed many non-PHP.B-like sequence families that enriched through selection for transduction of cells in the CNS.
  • AAV-PHP.C1 RYQGDSV
  • AAV-PHP.C2 WSTNAGY
  • AAV- PHP.C3 ERVGFAQ (FIG. 30). These showed enhanced BBB crossing irrespective of mouse strain, with roughly equal CNS transduction in BALB/cJ and C57BL/6J (FIGS. 32 and 73).
  • TQIPFK (SEQ ID: 435) is positioned between AA 588-589 of AAV capsid. The insertion sequence is underlined and bold.
  • Peptide sequence“DGTTLKPFLAQ” (SEQ ID: 867) is positioned by replacing AA 587- 590 of AAV capsid. The inserted sequence is underlined and highlighted.
  • a subject having Huntington's disease is identified.
  • the subject is then systemically administered a first amount of a viral vector that includes a polynucleotide that encodes for a Zinc finger protein (ZFP) engineered to represses the transcription of the Huntingtin (HTT) gene.
  • ZFP Zinc finger protein
  • the vector will be encapsidated by a modified AAV capsid protein with an amino acid sequence provided in FIG. 33 or provided in Tables 2-4, so as to allow proper targeting of the ZFP to the nervous system, among other organs.
  • the subject is administered a second or third dose of the vector, until a therapeutically effective amount of the ZFP is expressed in the subject in the nervous system.
  • a phase 1A clinical trial is performed to evaluate the safety, tolerability,
  • test composition comprising viral vector encapsidated by a modified capsid protein with any of the amino acid sequences provided in FIG. 33 or Tables 2-4, in subjects with late Huntington’s Disease (HD).
  • Eligible subjects are men and women between 21 and 65 years of age.
  • Inclusion Criteria Eligible subjects are men and women between 21 and 65 years of age. Subjects that (i) sign and date Sign and date International Classification of Functioning, Disability and Health (ICF); (2) male or female participant aged > 21 and ⁇ 65; (3) participants who submit medical report (PCR) attesting Huntington's disease with a number of CAG repeats on chromosome 4, greater than or equal to 40 and less than or equal to 50 (if the participant has not performed the examination and/or if he does not have the report available, a new exam should be done); (4) Score 5 points or more in motor assessment UHDRS scale (Unified
  • Exclusion Criteria (1) Any medical observation data (clinical and physical) that medical research judge as a risk for subject if enrollment at the study; (2) any laboratory exam data that medical research judge as a risk for subject if enrollment at the study; (4) history of epilepsy; (5) diagnostic of major cognitive impairment; (6) active decompensated psychiatric disease; (7) current or prior history of neoplasia; (8) current history of gastrointestinal, hepatic, renal, endocrine, pulmonary, hematologic, immune, metabolic pathology or severe and uncontrolled cardiovascular disease; (8) diagnostic of any active infection, be it viral, bacterial, fungal, or caused by another pathogen; (9) participants who have contraindication to undergo any of the tests performed in this study, for example, have pacemakers or surgical clip; (10) history of alcohol or illegal drugs abusers; (11) history of 1 or more episodes of suicide in the two years before Visit V-4; (12) active smoker or have stopped smoking less than six months prior to enrollment; (13) test positive in at least one of
  • Test High Dose One-time injection of test composition 2 x 10 A 10 vg at Week 0.
  • Test Middle Dose One-time injection of test composition 6 x 10 A 9 vg at Week 0.
  • Test Low Dose One-time injection of test composition 2x 10 A 9 vg at Week 0.
  • Test Lowest Dose One-time injection of test composition 2 x 10 A 8 vg at Week 0.
  • the immunological response induced by Cellavita HD will be evaluated by statistical comparison between baseline results of CD4+ and CD8+ proliferation and the other evaluated times.
  • Preliminary efficacy of Cellavita HD by comparison of the CNS assessment [ Time Frame: one year ].
  • Risk of suicidal ideation by Hamilton Depression Rating Scale (HDRS) [ Time Frame: five years ] will be evaluated by suicidal domain.
  • the classificatory pontuation may correspond to mild depression (score: 8 to 13), moderate depression (score: 19 - 22) and severe depression (score: > 23).

Abstract

La présente invention concerne des compositions et des kits comprenant des virus adéno-associés de recombinaison (VAAr) avec des tropismes qui présentent une spécificité et une efficacité accrues de transduction virale dans des types de cellules ciblées, par exemple, le cerveau et le foie. L'invention concerne également des utilisations thérapeutiques et biomédicales de recherche du VAAr, comprenant sans caractère limitatif des procédés de découverte de VAAr à l'aide d'un procédé d'évolution ciblée de VAA basée sur une recombinaison Cre multiplexée (M-CREATE) et des méthodes de traitement de diverses maladies et affections par une thérapie transgénique induite par VAAr.
PCT/US2020/027708 2019-04-11 2020-04-10 Compositions virales à spécificité améliorée dans le cerveau WO2020210655A1 (fr)

Priority Applications (8)

Application Number Priority Date Filing Date Title
AU2020271108A AU2020271108A1 (en) 2019-04-11 2020-04-10 Virus compositions with enhanced specificity in the brain
EP20787580.8A EP3953371A4 (fr) 2019-04-11 2020-04-10 Compositions virales à spécificité améliorée dans le cerveau
CN202080043549.2A CN114127090A (zh) 2019-04-11 2020-04-10 在大脑中具有增强的特异性的病毒组合物
CA3136658A CA3136658A1 (fr) 2019-04-11 2020-04-10 Compositions virales a specificite amelioree dans le cerveau
US17/602,505 US20220220502A1 (en) 2019-04-11 2020-04-10 Virus compositions with enhanced specificity in the brain
JP2021559829A JP2022526018A (ja) 2019-04-11 2020-04-10 脳内への特異性が増強されたウイルス組成物
KR1020217036564A KR20220007056A (ko) 2019-04-11 2020-04-10 뇌에서 증진된 특이성을 갖는 바이러스 조성물
SG11202111249YA SG11202111249YA (en) 2019-04-11 2020-04-10 Virus compositions with enhanced specificity in the brain

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962832826P 2019-04-11 2019-04-11
US62/832,826 2019-04-11

Publications (1)

Publication Number Publication Date
WO2020210655A1 true WO2020210655A1 (fr) 2020-10-15

Family

ID=72751358

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/027708 WO2020210655A1 (fr) 2019-04-11 2020-04-10 Compositions virales à spécificité améliorée dans le cerveau

Country Status (9)

Country Link
US (1) US20220220502A1 (fr)
EP (1) EP3953371A4 (fr)
JP (1) JP2022526018A (fr)
KR (1) KR20220007056A (fr)
CN (1) CN114127090A (fr)
AU (1) AU2020271108A1 (fr)
CA (1) CA3136658A1 (fr)
SG (1) SG11202111249YA (fr)
WO (1) WO2020210655A1 (fr)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11149256B2 (en) 2018-09-26 2021-10-19 California Institute Of Technology Adeno-associated virus compositions for targeted gene therapy
WO2022129609A3 (fr) * 2020-12-18 2022-09-15 Ac Immune Sa Administration d'anticorps
WO2022221421A3 (fr) * 2021-04-13 2022-11-17 Capsida, Inc. Compositions d'aav ayant une expression cérébrale élevée pour le traitement de la mucopolysaccharidose ii
WO2022221400A3 (fr) * 2021-04-13 2022-11-24 Capsida, Inc. Compositions aav ayant des niveaux d'expression élevés dans le cerveau
WO2022221420A3 (fr) * 2021-04-13 2022-11-24 Capsida, Inc. Compositions de vaa sélectionnées ayant un enrichissement cérébral préféré
US11518787B2 (en) 2018-07-11 2022-12-06 The Brigham And Women's Hospital, Inc. Methods and compositions for delivery of agents across the blood-brain barrier
WO2023004367A3 (fr) * 2021-07-20 2023-02-23 The Broad Institute, Inc. Compositions de ciblage modifiées pour cellules endothéliales du système vasculaire du système nerveux central et leurs procédés d'utilisation
WO2023091934A1 (fr) * 2021-11-16 2023-05-25 California Institute Of Technology Compositions de virus adéno-associés ayant un enrichissement de muscle cardiaque et squelettique préféré
WO2023148617A1 (fr) * 2022-02-01 2023-08-10 The Broad Institute, Inc. Vecteurs viraux adéno-associés et leurs utilisations
WO2023150632A3 (fr) * 2022-02-02 2023-09-28 The Broad Institute, Inc. Fractions de ciblage favorisant la transduction de cellules et de tissus du système nerveux central et procédés d'utilisation
US11820794B2 (en) 2019-11-22 2023-11-21 California Institute Of Technology Method for robust control of gene expression
WO2023235791A1 (fr) * 2022-06-02 2023-12-07 Voyager Therapeutics, Inc. Variants de capside de vaa et leurs utilisations
US11859200B2 (en) 2020-05-13 2024-01-02 Voyager Therapeutics, Inc. AAV capsids with increased tropism to brain tissue
WO2024025042A1 (fr) * 2022-07-26 2024-02-01 숙명여자대학교 산학협력단 Composition comprenant un virus recombinant aav2-anks1a pour le traitement de la maladie d'alzheimer
WO2024086747A1 (fr) 2022-10-19 2024-04-25 Affinia Therapeutics Inc. Aavs recombinants à tropisme et spécificité améliorés
US11981705B2 (en) 2021-01-08 2024-05-14 The Brigham And Women's Hospital, Inc. Methods and compositions for delivery of immunotherapy agents across the blood-brain barrier to treat brain cancer

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116693633B (zh) * 2023-02-21 2023-12-22 广州派真生物技术有限公司 腺相关病毒突变体及其应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150079038A1 (en) * 2013-09-13 2015-03-19 California Institute Of Technology Selective recovery
WO2019046069A1 (fr) * 2017-08-28 2019-03-07 The Regents Of The University Of California Variants de capside de virus adéno-associé et leurs procédés d'utilisation

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017100671A1 (fr) * 2015-12-11 2017-06-15 California Institute Of Technology Peptides de ciblage pour diriger des virus adéno-associés (aav)

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150079038A1 (en) * 2013-09-13 2015-03-19 California Institute Of Technology Selective recovery
WO2019046069A1 (fr) * 2017-08-28 2019-03-07 The Regents Of The University Of California Variants de capside de virus adéno-associé et leurs procédés d'utilisation

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3953371A4 *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11518787B2 (en) 2018-07-11 2022-12-06 The Brigham And Women's Hospital, Inc. Methods and compositions for delivery of agents across the blood-brain barrier
US11149256B2 (en) 2018-09-26 2021-10-19 California Institute Of Technology Adeno-associated virus compositions for targeted gene therapy
US11820794B2 (en) 2019-11-22 2023-11-21 California Institute Of Technology Method for robust control of gene expression
US11859200B2 (en) 2020-05-13 2024-01-02 Voyager Therapeutics, Inc. AAV capsids with increased tropism to brain tissue
WO2022129609A3 (fr) * 2020-12-18 2022-09-15 Ac Immune Sa Administration d'anticorps
US11981705B2 (en) 2021-01-08 2024-05-14 The Brigham And Women's Hospital, Inc. Methods and compositions for delivery of immunotherapy agents across the blood-brain barrier to treat brain cancer
WO2022221420A3 (fr) * 2021-04-13 2022-11-24 Capsida, Inc. Compositions de vaa sélectionnées ayant un enrichissement cérébral préféré
WO2022221400A3 (fr) * 2021-04-13 2022-11-24 Capsida, Inc. Compositions aav ayant des niveaux d'expression élevés dans le cerveau
WO2022221421A3 (fr) * 2021-04-13 2022-11-17 Capsida, Inc. Compositions d'aav ayant une expression cérébrale élevée pour le traitement de la mucopolysaccharidose ii
WO2023004367A3 (fr) * 2021-07-20 2023-02-23 The Broad Institute, Inc. Compositions de ciblage modifiées pour cellules endothéliales du système vasculaire du système nerveux central et leurs procédés d'utilisation
WO2023091934A1 (fr) * 2021-11-16 2023-05-25 California Institute Of Technology Compositions de virus adéno-associés ayant un enrichissement de muscle cardiaque et squelettique préféré
WO2023148617A1 (fr) * 2022-02-01 2023-08-10 The Broad Institute, Inc. Vecteurs viraux adéno-associés et leurs utilisations
WO2023150632A3 (fr) * 2022-02-02 2023-09-28 The Broad Institute, Inc. Fractions de ciblage favorisant la transduction de cellules et de tissus du système nerveux central et procédés d'utilisation
WO2023235791A1 (fr) * 2022-06-02 2023-12-07 Voyager Therapeutics, Inc. Variants de capside de vaa et leurs utilisations
WO2024025042A1 (fr) * 2022-07-26 2024-02-01 숙명여자대학교 산학협력단 Composition comprenant un virus recombinant aav2-anks1a pour le traitement de la maladie d'alzheimer
WO2024086747A1 (fr) 2022-10-19 2024-04-25 Affinia Therapeutics Inc. Aavs recombinants à tropisme et spécificité améliorés

Also Published As

Publication number Publication date
CN114127090A (zh) 2022-03-01
JP2022526018A (ja) 2022-05-20
EP3953371A1 (fr) 2022-02-16
US20220220502A1 (en) 2022-07-14
KR20220007056A (ko) 2022-01-18
AU2020271108A1 (en) 2021-11-11
SG11202111249YA (en) 2021-11-29
CA3136658A1 (fr) 2020-10-15
EP3953371A4 (fr) 2023-04-12

Similar Documents

Publication Publication Date Title
US20220220502A1 (en) Virus compositions with enhanced specificity in the brain
US11149256B2 (en) Adeno-associated virus compositions for targeted gene therapy
US20230131352A1 (en) Redirection of tropism of aav capsids
US20230303630A1 (en) Adeno-associated virus compositions having preferred expression levels
EP3861010A1 (fr) Redirection de tropisme de capsides aav
WO2022221404A2 (fr) Compositions de virus adéno-associés ayant un enrichissement cérébral accru
US20240150410A1 (en) Aav compositions having high expression levels in brain
WO2022221420A2 (fr) Compositions de vaa sélectionnées ayant un enrichissement cérébral préféré
EP4323015A1 (fr) Virions de vaa à polyploïde rationnel traversant la barrière hémato-encéphalique et déclenchant une réponse humorale réduite
WO2023225508A2 (fr) Compositions de virus adéno-associé sélectionnées ayant des niveaux d'expression cérébrale, médullaire et/ou cardiaque préférés
WO2023215546A2 (fr) Compositions de virus adéno-associés se caractérisant par un enrichissement cérébral accru
WO2024086628A2 (fr) Compositions de virus adéno-associés ayant un enrichissement cérébral préféré et un enrichissement hépatique faible
WO2023183582A2 (fr) Compositions de vaa sélectionnés ayant un enrichissement cérébral préféré
WO2023244919A1 (fr) Compositions de virus adéno-associé ayant un enrichissement cérébral accru et un enrichissement hépatique réduit
WO2023244920A2 (fr) Compositions de virus adéno-associé ayant un enrichissement cérébral et/ou un enrichissement cardiaque accrus

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 20787580

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2021559829

Country of ref document: JP

Kind code of ref document: A

Ref document number: 3136658

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2020271108

Country of ref document: AU

Date of ref document: 20200410

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2020787580

Country of ref document: EP

Effective date: 20211111